1
|
Yoshimura S, Dorok M, Mamrak U, Wehn A, Krestel E, Khalin I, Plesnila N. Reliable infarction of the middle cerebral artery territory in C57BL/6 mice using pterygopalatine artery ligation and filament optimization - The PURE-MCAo model. J Cereb Blood Flow Metab 2025; 45:871-884. [PMID: 39370987 PMCID: PMC11563556 DOI: 10.1177/0271678x241281841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/30/2024] [Accepted: 08/22/2024] [Indexed: 10/08/2024]
Abstract
Current techniques for inducing intraluminal filamentous middle cerebral artery occlusion (fMCAo) in mice produce highly variable results and often cause additional infarcts in the posterior cerebral artery (PCA) territory. The aim of the current study was to develop a novel procedure to overcome these shortcomings. Male C57BL/6 mice were subjected to 60 min of fMCAo with cerebral blood flow monitored by laser Doppler flowmetry. The influence of the length of the occlusion filament coating and the combination of common carotid artery (CCA) or pterygopalatine artery (PPA) ligation on lesion volume and functional outcome 24 h after reperfusion was evaluated. The use of appropriate filament and PPA ligation while maintaining CCA perfusion prevented the development of infarcts in the PCA area, resulted in pure MCA infarcts (68.3 ± 14.5 mm3) and reduced the variability of infarct volumes by more than half (from 26-38% to 14% standard deviation/mean). Using an improved fMCAo procedure, we were able to produce PCA area-unaffected reproducible (PURE) infarcts exclusively in the MCA territory. Thus PURE-MCAo reduced outcome variability by more than 50%. Our results may thus help to reduce the number of animals in preclinical stroke research and to increase the reproducibility of the fMCAo model.
Collapse
Affiliation(s)
- Sodai Yoshimura
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Department of Neurosurgery, Nihon University School of Medicine, Tokyo, Japan
| | - Maximilian Dorok
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
| | - Uta Mamrak
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
| | - Antonia Wehn
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Department of Neurosurgery, LMU University Hospital, Munich, Germany
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| |
Collapse
|
2
|
Faber JE. Collateral blood vessels in stroke and ischemic disease: Formation, physiology, rarefaction, remodeling. J Cereb Blood Flow Metab 2025:271678X251322378. [PMID: 40072222 PMCID: PMC11904929 DOI: 10.1177/0271678x251322378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Collateral blood vessels are unique, naturally occurring endogenous bypass vessels that provide alternative pathways for oxygen delivery in obstructive arterial conditions and diseases. Surprisingly however, the capacity of the collateral circulation to provide protection varies greatly among individuals, resulting in a significant fraction having poor collateral circulation in their tissues. We recently reviewed evidence that the presence of naturally-occurring polymorphisms in genes that determine the number and diameter of collaterals that form during development (ie, genetic background), is a major contributor to this variation. The purpose of this review is to summarize current understanding of the other determinants of collateral blood flow, drawing on both animal and human studies. These include the level of smooth muscle tone in collaterals, hemodynamic forces, how collaterals form during development (collaterogenesis), de novo formation of additional new collaterals during adulthood, loss of collaterals with aging and cardiovascular risk factor presence (rarefaction), and collateral remodeling (structural lumen enlargement). We also review emerging evidence that collaterals not only provide protection in ischemic conditions but may also serve a physiological function in healthy individuals. Primary focus is on studies conducted in brain, however relevant findings in other tissues are also reviewed, as are questions for future investigation.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, Curriculum in Neuroscience, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
3
|
Törteli A, Tóth R, Bari F, Farkas E, Menyhárt Á. Collateral is brain: Low perfusion triggers spreading depolarization and futile reperfusion after acute ischemic stroke. J Cereb Blood Flow Metab 2024; 44:1881-1887. [PMID: 39225037 PMCID: PMC11529658 DOI: 10.1177/0271678x241270480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/30/2024] [Indexed: 09/04/2024]
Abstract
Futile reperfusion is a phenomenon of inadequate perfusion despite successful recanalization after acute ischemic stroke (AIS). It is associated with poor patient outcomes and has received increasing interest due to its clinical diagnosis becoming more common. However, the underlying mechanisms remain elusive, and experimental studies are focused on the pathological background of futile reperfusion. Our recent study has confirmed that poor primary collateralization plays a crucial role in the insufficiency of reperfusion after AIS in mice. Specifically, the absence of primary collaterals in the circle of Willis (CoW) promoted the development of spreading depolarizations (SDs) during AIS. In our experimental stroke model, the occurrence of SDs during ischemia always predicted futile reperfusion. Conversely, in mice with a complete CoW, no SDs were observed, and reperfusion was complete. Importantly, the human CoW displays variation in the primary collaterals in approximately 50% of the population. Therefore, futile reperfusion may result from SD evolution in AIS patients. Our purpose here is to emphasize the crucial role of SD in the development of futile reperfusion. We propose that adequate collateral recruitment can prevent SD occurrence, leading to improved reperfusion and AIS outcomes.
Collapse
Affiliation(s)
- Anna Törteli
- Hungarian Centre of Excellence for Molecular Medicine – University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Réka Tóth
- Hungarian Centre of Excellence for Molecular Medicine – University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine – University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine – University of Szeged Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
4
|
Zdunczyk A, Schumm L, Helgers SOA, Nieminen-Kelhä M, Bai X, Major S, Dreier JP, Hecht N, Woitzik J. Ketamine-induced prevention of SD-associated late infarct progression in experimental ischemia. Sci Rep 2024; 14:10186. [PMID: 38702377 PMCID: PMC11068759 DOI: 10.1038/s41598-024-59835-5] [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: 01/04/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024] Open
Abstract
Spreading depolarizations (SDs) occur frequently in patients with malignant hemispheric stroke. In animal-based experiments, SDs have been shown to cause secondary neuronal damage and infarct expansion during the initial period of infarct progression. In contrast, the influence of SDs during the delayed period is not well characterized yet. Here, we analyzed the impact of SDs in the delayed phase after cerebral ischemia and the potential protective effect of ketamine. Focal ischemia was induced by distal occlusion of the left middle cerebral artery in C57BL6/J mice. 24 h after occlusion, SDs were measured using electrocorticography and laser-speckle imaging in three different study groups: control group without SD induction, SD induction with potassium chloride, and SD induction with potassium chloride and ketamine administration. Infarct progression was evaluated by sequential MRI scans. 24 h after occlusion, we observed spontaneous SDs with a rate of 0.33 SDs/hour which increased during potassium chloride application (3.37 SDs/hour). The analysis of the neurovascular coupling revealed prolonged hypoemic and hyperemic responses in this group. Stroke volume increased even 24 h after stroke onset in the SD-group. Ketamine treatment caused a lesser pronounced hypoemic response and prevented infarct growth in the delayed phase after experimental ischemia. Induction of SDs with potassium chloride was significantly associated with stroke progression even 24 h after stroke onset. Therefore, SD might be a significant contributor to delayed stroke progression. Ketamine might be a possible drug to prevent SD-induced delayed stroke progression.
Collapse
Affiliation(s)
- A Zdunczyk
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - L Schumm
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - S O A Helgers
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - M Nieminen-Kelhä
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - X Bai
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - S Major
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - J P Dreier
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - N Hecht
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
- Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
- University Clinic for Neurosurgery, Marienstr. 11, 26121, Oldenburg, Germany.
| |
Collapse
|
5
|
Zhang L, Wong LR, Wong P, Shen W, Yang S, Huang L, Lim YA, Ho PCL. Chronic treatment with baicalein alleviates behavioural disorders and improves cerebral blood flow via reverting metabolic abnormalities in a J20 transgenic mouse model of Alzheimer's disease. Brain Behav Immun Health 2023; 28:100599. [PMID: 36817510 PMCID: PMC9931920 DOI: 10.1016/j.bbih.2023.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
Baicalein (BE) has both antioxidant and anti-inflammatory effects. It has also been reported able to improve cerebral blood circulation in brain ischemic injury. However, its chronic efficacy and metabolomics in Alzheimer's disease (AD) remain unknown. In this study, BE at 80 mg/kg was administrated through the oral route in J20 AD transgenic mice aged from aged 4 months to aged 10 months. Metabolic- and neurobehavioural phenotyping was done before and after 6 months' treatment to evaluate the drug efficacy and the relevant mechanisms. Meanwhile, molecular docking was used to study the binding affinity of BE and poly (ADP-ribose) polymerase-1 (PARP-1) which is related to neuronal injury. The open field test showed that BE could suppress hyperactivity in J20 mice and increase the frequency of the target quadrant crossing in the Morris Water Maze test. More importantly, BE restored cerebral blood flow back to the normal level after the chronic treatment. A 1H NMR-based metabolomics study showed that BE treatment could restore the tricarboxylic acid cycle in plasma. And such a treatment could suppress oxidative stress, inhibit neuroinflammation, alleviate mitochondrial dysfunction, improve neurotransmission, and restore amino homeostasis via starch and sucrose metabolism and glycolipid metabolism in the cortex and hippocampus, which could affect the behavioural and cerebral blood flow. These findings showed that BE is a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Li Zhang
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 117583, Singapore,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Ling Rong Wong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Peiyan Wong
- Neuroscience Phenotyping Core, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Wanxiang Shen
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Shili Yang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yun-An Lim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Paul Chi-Lui Ho
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 117583, Singapore,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore,Monash University Malaysia, School of Pharmacy, Subang Jaya, 47500, Selangor, Malaysia,Corresponding author. Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
| |
Collapse
|
6
|
Törteli A, Tóth R, Berger S, Samardzic S, Bari F, Menyhárt Á, Farkas E. Spreading depolarization causes reperfusion failure after cerebral ischemia. J Cereb Blood Flow Metab 2023; 43:655-664. [PMID: 36703609 PMCID: PMC10108181 DOI: 10.1177/0271678x231153745] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Despite successful recanalization, reperfusion failure associated with poor neurological outcomes develops in half of treated stroke patients. We explore here whether spreading depolarization (SD) is a predictor of reperfusion failure. Global forebrain ischemia/reperfusion was induced in male and female C57BL/6 mice (n = 57). SD and cerebral blood flow (CBF) changes were visualized with transcranial intrinsic optical signal and laser speckle contrast imaging. To block SD, MK801 was applied (n = 26). Neurological deficit, circle of Willis (CoW) anatomy and neuronal injury were evaluated 24 hours later. SD emerged after ischemia onset in one or both hemispheres under a perfusion threshold (CBF drop to 21.1 ± 4.6 vs. 33.6 ± 4.4%, SD vs. no SD). The failure of later reperfusion (44.4 ± 12.5%) was invariably linked to previous SD. In contrast, reperfusion was adequate (98.9 ± 7.4%) in hemispheres devoid of SD. Absence of the P1 segment of the posterior cerebral artery in the CoW favored SD occurrence and reperfusion failure. SD occurrence and reperfusion failure were associated with poor neurologic function, and neuronal necrosis 24 hours after ischemia. The inhibition of SD significantly improved reperfusion. SD occurrence during ischemia impairs later reperfusion, prognosticating poor neurological outcomes. The increased likelihood of SD occurrence is predicted by inadequate collaterals.
Collapse
Affiliation(s)
- Anna Törteli
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Réka Tóth
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Sarah Berger
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Sarah Samardzic
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ákos Menyhárt
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged, Cerebral Blood Flow and Metabolism Research Group, Szeged, Hungary.,Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
7
|
Pi C, Wang J, Zhao D, Yu S. The determinants of collateral circulation status in patients with chronic cerebral arterial circle occlusion: A STROBE Study. Medicine (Baltimore) 2022; 101:e29703. [PMID: 35777030 PMCID: PMC9239625 DOI: 10.1097/md.0000000000029703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The condition of collateral pathways is an important predictor of stroke prognoses; however the major determinants of collaterals are still unknown. The purpose of this study is to identify potentially determinants for collateral circulation status in patients with chronic occlusion of cerebral arterial circle. All patients with chronic occlusion of either unilateral internal carotid artery or middle cerebral artery M1 or M2 segment, diagnosed by digital subtraction angiography at the neurology department of the First Medical Centre of Chinese PLA General Hospital from January 2015 to December 2017, were retrospectively collected in our sample. After screening according to inclusion and exclusion criteria, the patients' relevant clinical data were collected and analyzed. Collateral circulations were assessed by 2 independent raters using the American society of interventional and therapeutic neuroradiology/society of interventional radiology flow-grading system. Baseline characteristics (n = 163): our sample consists of 116 (71.2%) male and 47 (28.8%) female patients with an average age of 57.5 ± 11.9 years. Cerebral collateral flow was poor in 59 (36.2%) patients. Our univariate analyses showed that poor collateral circulation was associated with lower high-density lipoproteins cholesterol (HDL), elevated homocysteine levels, aging and hyperlipidemia. A multivariate analysis identified HDL, homocysteine levels and ageing as major predictors for collateral circulation status. In the subgroup analysis, the HDL contributed to collateral angiogenesis internal carotid artery occlusion group. In the middle cerebral artery occlusion group, the homocysteine and ageing were related to the poor collateral status. Low HDL, high levels of homocysteine and ageing are identified as possible risk factors for a poor collateral vessel blood flow in patients with chronic anterior circulation occlusion.
Collapse
Affiliation(s)
- Chenghui Pi
- Nankai University, College of Medicine, Tianjin, China
- Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jun Wang
- Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Dengfa Zhao
- Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Shengyuan Yu
- Nankai University, College of Medicine, Tianjin, China
- Department of Neurology, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Shengyuan Yu, College of Medicine, Nankai University, Tianjin, China )
| |
Collapse
|
8
|
Xue Y, Zeng C, Ge P, Liu C, Li J, Zhang Y, Zhang D, Zhang Q, Zhao J. Association of RNF213 Variants With Periventricular Anastomosis in Moyamoya Disease. Stroke 2022; 53:2906-2916. [PMID: 35543128 DOI: 10.1161/strokeaha.121.038066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The pathogenic mechanisms of periventricular anastomosis (PA) in moyamoya disease remain unknown. Here, we aimed to describe the angiographic profiles of PA and their relationships with really interesting new gene (RING) finger protein 213 (RNF213) genotypes. METHODS We conducted a retrospective cohort study of moyamoya disease patients consecutively recruited between June 2019 and January 2021 in Beijing Tiantan Hospital, Capital Medical University, China. C-terminal region of RNF213 was sequenced. Angiographic characteristics of PA vessels (lenticulostriate artery, thalamotuberal artery, thalamoperforating artery, anterior choroidal artery, and posterior choroidal artery) were compared between different groups of RNF213 genotypes. The dilatation and extension of PA vessels were measured by using PA score (positive, score 1-5; negative, score 0). Multivariate regression analysis was conducted to assess variables associated with PA score. In addition, gene expression of RNF213 in human brain regions was evaluated from the Allen Human Brain Atlas. RESULTS Among 260 patients (484 hemispheres), 71.2% carried no RNF213 rare and novel variants, 20.0% carried p.R4810K heterozygotes, and 8.8% carried other rare and novel variants. PA scores in patients with p.R4810K and other rare and novel variants were significantly higher than in wild-type patients (P<0.001). Age (odds ratio [OR], 0.958 [95% CI, 0.942-0.974]; P<0.001), platelet count (OR, 0.996 [95% CI, 0.992-0.999]; P=0.027), p.R4810K variant (OR, 2.653 [95% CI, 1.514-4.649]; P=0.001), other rare and novel variants (OR, 3.197 [95% CI, 1.012-10.094]; P=0.048), Suzuki stage ≥4 (OR, 1.941 [95% CI, 1.138-3.309]; P=0.015), and posterior cerebral artery involvement (OR, 1.827 [95% CI, 1.020-3.271]; P=0.043) were significantly correlated with PA score. High expression of RNF213 was detected in the periventricular area. CONCLUSIONS RNF213 variants were confirmed to be associated with PA in moyamoya disease. Individuals with RNF213 p.R4810K heterozygotes and other C-terminal region rare variants exhibited different angiographic phenotypes, compared with wild-type patients.
Collapse
Affiliation(s)
- Yimeng Xue
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing (Y.X., J.Z.).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Chaofan Zeng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| | - Jizong Zhao
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing (Y.X., J.Z.).,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,China National Clinical Research Center for Neurological Diseases, Beijing (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Center of Stroke, Beijing Institute for Brain Disorders, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.).,Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, China (Y.X., C.Z., P.G., C.L., J.L., Y.Z., D.Z., Q.Z., J.Z.)
| |
Collapse
|
9
|
Luo B, Liu J, Xiong L, Fang C, He Y. Normal cerebral blood vessels under ultrasound in SD rats of different ages. IBRAIN 2022; 8:346-352. [PMID: 37786747 PMCID: PMC10528998 DOI: 10.1002/ibra.12035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 10/04/2023]
Abstract
The objective of this study was to examine whether ultrasound can examine the development of cerebral vascular structure and cerebral blood flow in Sprague-Dawley (SD) rats by ultrasound in a noninvasive manner, which provides a reference for ultrasound research of SD rats. Thirty-nine SD rats (7-16 days old) were divided into seven groups according to age, and the number of SD rats in each group was, respectively, 7, 17, 1, 3, 2, 8, and 1. Ultrasound was used to detect cerebral blood vessels, cerebrovascular flow velocity, and heart rate in SD rats in the sagittal and coronal positions, and images were obtained in B-mode ultrasound. The cerebral vascular structure of 39 SD rats (7-16 days) was dynamically observed under B-ultrasound. We found that the cerebral vascular structure of the rats aged 7-10 days was clear and detectable. Rats aged 11-16 days of cerebral vascular structures became thinner and undetectable. Quantitative analysis of cerebrovascular flow rate and heart rate in rats found that there was no significant difference in cerebrovascular blood flow rate and heart rate between 7 and 8 days. Ultrasound can also be used in rat animal studies, that is, the cerebral blood flow in rats of different ages can be monitored in real-time by ultrasound in a noninvasive way.
Collapse
Affiliation(s)
- Bo‐Yan Luo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of PharmacyZunyi Medical UniversityZunyiGuizhouChina
| | - Jin‐Xiang Liu
- Animal Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Liu‐Lin Xiong
- Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Chang‐Le Fang
- School of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Yu‐Qi He
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of PharmacyZunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
10
|
Kaloss AM, Theus MH. Leptomeningeal anastomoses: Mechanisms of pial collateral remodeling in ischemic stroke. WIREs Mech Dis 2022; 14:e1553. [PMID: 35118835 PMCID: PMC9283306 DOI: 10.1002/wsbm.1553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Abstract
Arterial collateralization, as determined by leptomeningeal anastomoses or pial collateral vessels, is a well‐established vital player in cerebral blood flow restoration and neurological recovery from ischemic stroke. A secondary network of cerebral collateral circulation apart from the Circle of Willis, exist as remnants of arteriole development that connect the distal arteries in the pia mater. Recent interest lies in understanding the cellular and molecular adaptations that control the growth and remodeling, or arteriogenesis, of these pre‐existing collateral vessels. New findings from both animal models and human studies of ischemic stroke suggest a multi‐factorial and complex, temporospatial interplay of endothelium, immune and vessel‐associated cell interactions may work in concert to facilitate or thwart arteriogenesis. These valuable reports may provide critical insight into potential predictors of the pial collateral response in patients with large vessel occlusion and may aid in therapeutics to enhance collateral function and improve recovery from stroke. This article is categorized under:Neurological Diseases > Molecular and Cellular Physiology
Collapse
Affiliation(s)
- Alexandra M Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA.,School of Neuroscience, Virginia Tech, Blacksburg, Virginia, USA.,Center for Regenerative Medicine, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
11
|
Schumm L, Lemale CL, Major S, Hecht N, Nieminen-Kelhä M, Zdunczyk A, Kowoll CM, Martus P, Thiel CM, Dreier JP, Woitzik J. Physiological variables in association with spreading depolarizations in the late phase of ischemic stroke. J Cereb Blood Flow Metab 2022; 42:121-135. [PMID: 34427143 PMCID: PMC8721769 DOI: 10.1177/0271678x211039628] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Physiological effects of spreading depolarizations (SD) are only well studied in the first hours after experimental stroke. In patients with malignant hemispheric stroke (MHS), monitoring of SDs is restricted to the postoperative ICU stay, typically day 2-7 post-ictus. Therefore, we investigated the role of physiological variables (temperature, intracranial pressure, mean arterial pressure and cerebral perfusion pressure) in relationship to SD during the late phase after MHS in humans. Additionally, an experimental stroke model was used to investigate hemodynamic consequences of SD during this time window. In 60 patients with MHS, the occurrence of 1692 SDs was preceded by a decrease in mean arterial pressure (-1.04 mmHg; p = .02) and cerebral perfusion pressure (-1.04 mmHg; p = .03). Twenty-four hours after middle cerebral artery occlusion in 50 C57Bl6/J mice, hypothermia led to prolonged SD-induced hyperperfusion (+2.8 min; p < .05) whereas hypertension mitigated initial hypoperfusion (-1.4 min and +18.5%Δ rCBF; p < .01). MRI revealed that SDs elicited 24 hours after experimental stroke were associated with lesion progression (15.9 vs. 14.8 mm³; p < .01). These findings of small but significant effects of physiological variables on SDs in the late phase after ischemia support the hypothesis that the impact of SDs may be modified by adjusting physiological variables.
Collapse
Affiliation(s)
- Leonie Schumm
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurosurgery, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Coline L Lemale
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Major
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Zdunczyk
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Peter Martus
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute for Clinical Epidemiology and Applied Biostatistics, University of Tübingen, Tübingen, Germany
| | - Christiane M Thiel
- Biological Psychology, Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Jens P Dreier
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
12
|
de Bortoli T, Boehm-Sturm P, Koch SP, Nieminen-Kelhä M, Wessels L, Mueller S, Ielacqua GD, Klohs J, Vajkoczy P, Hecht N. Three-Dimensional Iron Oxide Nanoparticle-Based Contrast-Enhanced Magnetic Resonance Imaging for Characterization of Cerebral Arteriogenesis in the Mouse Neocortex. Front Neurosci 2021; 15:756577. [PMID: 34899163 PMCID: PMC8662986 DOI: 10.3389/fnins.2021.756577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: Subsurface blood vessels in the cerebral cortex have been identified as a bottleneck in cerebral perfusion with the potential for collateral remodeling. However, valid techniques for non-invasive, longitudinal characterization of neocortical microvessels are still lacking. In this study, we validated contrast-enhanced magnetic resonance imaging (CE-MRI) for in vivo characterization of vascular changes in a model of spontaneous collateral outgrowth following chronic cerebral hypoperfusion. Methods: C57BL/6J mice were randomly assigned to unilateral internal carotid artery occlusion or sham surgery and after 21 days, CE-MRI based on T2*-weighted imaging was performed using ultra-small superparamagnetic iron oxide nanoparticles to obtain subtraction angiographies and steady-state cerebral blood volume (ss-CBV) maps. First pass dynamic susceptibility contrast MRI (DSC-MRI) was performed for internal validation of ss-CBV. Further validation at the histological level was provided by ex vivo serial two-photon tomography (STP). Results: Qualitatively, an increase in vessel density was observed on CE-MRI subtraction angiographies following occlusion; however, a quantitative vessel tracing analysis was prone to errors in our model. Measurements of ss-CBV reliably identified an increase in cortical vasculature, validated by DSC-MRI and STP. Conclusion: Iron oxide nanoparticle-based ss-CBV serves as a robust, non-invasive imaging surrogate marker for neocortical vessels, with the potential to reduce and refine preclinical models targeting the development and outgrowth of cerebral collateralization.
Collapse
Affiliation(s)
- Till de Bortoli
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Philipp Boehm-Sturm
- Center for Stroke Research Berlin (CSB), Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan P Koch
- Center for Stroke Research Berlin (CSB), Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Lars Wessels
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Susanne Mueller
- Center for Stroke Research Berlin (CSB), Berlin, Germany.,Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Giovanna D Ielacqua
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zürich, Zurich, Switzerland
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Center for Stroke Research Berlin (CSB), Berlin, Germany
| |
Collapse
|
13
|
Moore EE, Liu D, Bown CW, Kresge HA, Gupta DK, Pechman KR, Mendes LA, Davis LT, Gifford KA, Anderson AW, Wang TJ, Landman BA, Hohman TJ, Jefferson AL. Lower cardiac output is associated with neurodegeneration among older adults with normal cognition but not mild cognitive impairment. Brain Imaging Behav 2021; 15:2040-2050. [PMID: 33040257 PMCID: PMC8035362 DOI: 10.1007/s11682-020-00398-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2020] [Indexed: 01/21/2023]
Abstract
Subclinical cardiac dysfunction is associated with smaller total brain volume on magnetic resonance imaging (MRI). To study whether cardiac output relates to regional measurements of grey and white matter structure, older adults (n = 326) underwent echocardiogram to quantify cardiac output (L/min) and brain MRI. Linear regressions related cardiac output to grey matter volumes measured on T1 and white matter hyperintensities assessed on T2-FLAIR. Voxelwise analyses related cardiac output to diffusion tensor imaging adjusting for demographic, genetic, and vascular risk factors. Follow-up models assessed a cardiac output x diagnosis interaction with stratification (normal cognition, mild cognitive impairment). Cardiac output interacted with diagnosis, such that lower cardiac output related to smaller total grey matter (p = 0.01), frontal lobe (p = 0.01), and occipital lobe volumes (p = 0.01) among participants with normal cognition. When excluding participants with cardiovascular disease and atrial fibrillation, associations emerged with smaller parietal lobe (p = 0.005) and hippocampal volume (p = 0.05). Subtle age-related cardiac changes may disrupt neuronal homeostasis and impact grey matter integrity prior to cognitive impairment.
Collapse
Affiliation(s)
- Elizabeth E Moore
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
- Medical Scientist Training Program, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Dandan Liu
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Corey W Bown
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
| | - Hailey A Kresge
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
| | - Deepak K Gupta
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kimberly R Pechman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa A Mendes
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - L Taylor Davis
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine A Gifford
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam W Anderson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Thomas J Wang
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bennett A Landman
- Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela L Jefferson
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, 1207 17th Avenue South, Suite 204, Nashville, TN, 37212, USA.
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
14
|
Brothers RO, Atlas N, Cowdrick KR, Buckley EM. Cerebrovascular reactivity measured in awake mice using diffuse correlation spectroscopy. NEUROPHOTONICS 2021; 8:015007. [PMID: 33665230 PMCID: PMC7920384 DOI: 10.1117/1.nph.8.1.015007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/15/2021] [Indexed: 05/31/2023]
Abstract
Significance: Cerebrovascular reactivity (CVR), defined as the ability of the cerebral vasculature to dilate or constrict in response to a vasoactive stimulus, is an important indicator of the brain's vascular health. However, mechanisms of cerebrovascular dysregulation are poorly understood, and no effective treatment strategies for impaired CVR exist. Preclinical murine models provide an excellent platform for interrogating mechanisms underlying CVR dysregulation and determining novel therapeutics that restore impaired CVR. However, quantification of CVR in mice is challenging. Aim: We present means of assessing CVR in awake mice using intraperitoneal injection of acetazolamide (ACZ) combined with continuous monitoring of cerebral blood flow. Approach: Measurements of cerebral blood flow were made with a minimally invasive diffuse correlation spectroscopy sensor that was secured to an optical window glued to the intact skull. Two source-detector separations (3 and 4.5 mm) per hemisphere were used to probe different depths. CVR was quantified as the relative increase in blood flow due to ACZ. CVR was assessed once daily for 5 days in 5 mice. Results: We found that CVR and the response half-time were remarkably similar across hemispheres and across 3- versus 4.5-mm separations, suggesting a homogenous, whole brain response to ACZ. Mean(std) intra- and intermouse coefficients of variations were 15(9)% and 19(10)%, respectively, for global CVR and 24(15)% and 27(11)%, respectively, for global response half-time. Conclusion: In sum, we report a repeatable method of measuring CVR in free-behaving mice which can be used to screen for impairments with disease and to track changes in CVR with therapeutic interventions.
Collapse
Affiliation(s)
- Rowan O. Brothers
- Emory University and Georgia Institute of Technology, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Nir Atlas
- Emory University and Children’s Healthcare of Atlanta, Division of Critical Care Medicine, Department of Pediatrics, Atlanta, Georgia, United States
| | - Kyle R. Cowdrick
- Emory University and Georgia Institute of Technology, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Erin M. Buckley
- Emory University and Georgia Institute of Technology, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Emory University School of Medicine, Department of Pediatrics, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Children’s Research Scholar, Atlanta, Georgia, United States
| |
Collapse
|
15
|
Zhang H, Rzechorzek W, Aghajanian A, Faber JE. Hypoxia induces de novo formation of cerebral collaterals and lessens the severity of ischemic stroke. J Cereb Blood Flow Metab 2020; 40:1806-1822. [PMID: 32423327 PMCID: PMC7430105 DOI: 10.1177/0271678x20924107] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Pial collaterals provide protection in stroke. Evidence suggests their formation late during gestation (collaterogenesis) is driven by reduced oxygen levels in the cerebral watersheds. The purpose of this study was to determine if collaterogenesis can be re-activated in the adult to induce formation of additional collaterals ("neo-collateral formation", NCF). Mice were gradually acclimated to reduced inspired oxygen (FIO2) and maintained at 12, 10, 8.5 or 7% for two-to-eight weeks. Hypoxemia induced "dose"-dependent NCF and remodeling of native collaterals, and decreased infarct volume after permanent MCA occlusion. In contrast, no formation occurred of addition collateral-like intra-tree anastomoses, PComs, or branches within the MCA tree. Hypoxic NCF, remodeling and infarct protection were durable, i.e. retained for at least six weeks after return to normoxia. Hypoxia increased expression of Hif2α, Vegfa, Rabep2, Angpt2, Tie2 and Cxcr4. Neo-collateral formation was abolished in mice lacking Rabep2, a novel gene involved in VEGFA→Flk1 signaling and required for formation of collaterals during development, and inhibited by knockdown of Vegfa, Flk1 and Cxcr4. Rabep2-dependent NCF was also induced by permanent MCA occlusion. This is the first report that hypoxia induces new pial collaterals to form. Hypoxia- and occlusion-induced neo-collateral formation provide models to study collaterogenesis in the adult.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - Wojciech Rzechorzek
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| | - James E Faber
- Department of Cell Biology and Physiology, McAllister Heart Institute, Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
16
|
Pál É, Hricisák L, Lékai Á, Nagy D, Fülöp Á, Erben RG, Várbíró S, Sándor P, Benyó Z. Ablation of Vitamin D Signaling Compromises Cerebrovascular Adaptation to Carotid Artery Occlusion in Mice. Cells 2020; 9:cells9061457. [PMID: 32545499 PMCID: PMC7349396 DOI: 10.3390/cells9061457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Vitamin D insufficiency has been associated with increased incidence and severity of cerebrovascular disorders. We analyzed the impact of impaired vitamin D signaling on the anatomical and functional aspects of cerebrovascular adaptation to unilateral carotid artery occlusion (CAO), a common consequence of atherosclerosis and cause of ischemic stroke. Cerebrocortical blood flow (CoBF) showed a significantly increased drop and delayed recovery after CAO in mice carrying a functionally inactive vitamin D receptor (VDR) with the most sustained perfusion deficit in the temporal cortex. To identify the cause(s) for this altered adaptation, the extent of compensatory blood flow increase in the contralateral carotid artery and the morphology of pial collaterals between the anterior and middle cerebral arteries were determined. Whereas VDR deficiency had no significant influence on the contralateral carotid arterial blood flow increase, it was associated with decreased number and increased tortuosity of pial anastomoses resulting in unfavorable changes of the intracranial collateral circulation. These results indicate that VDR deficiency compromises the cerebrovascular adaptation to CAO with the most sustained consequences in the temporal cortex. The dysregulation can be attributed to the altered development and function of pial collateral circulation whereas extracranial vessels may not be impaired.
Collapse
Affiliation(s)
- Éva Pál
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
- Correspondence: (É.P.); (Z.B.); Tel.: +36-1-210-0306 (É.P.); +36-1-210-0306 (Z.B.)
| | - László Hricisák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
| | - Ágnes Lékai
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
| | - Dorina Nagy
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
| | - Ágnes Fülöp
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
| | - Reinhold G. Erben
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary;
| | - Péter Sándor
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (Á.L.); (D.N.); (Á.F.); (P.S.)
- Correspondence: (É.P.); (Z.B.); Tel.: +36-1-210-0306 (É.P.); +36-1-210-0306 (Z.B.)
| |
Collapse
|
17
|
Lee JS, Hong JM, Yoon BS, Son KS, Lee KE, Im DS, Park BN, An YS, Hwang DH, Park CB, Kim BG, Joe EH. Expression of Cellular Receptors in the Ischemic Hemisphere of Mice with Increased Glucose Uptake. Exp Neurobiol 2020; 29:70-79. [PMID: 32122109 PMCID: PMC7075656 DOI: 10.5607/en.2020.29.1.70] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/16/2020] [Accepted: 02/06/2020] [Indexed: 11/19/2022] Open
Abstract
Many previous studies have shown reduced glucose uptake in the ischemic brain. In contrast, in a permanent unilateral common carotid artery occlusion (UCCAO) mouse model, our pilot experiments using 18F-fluorodeoxyglucose positron emission tomography (FDG PET) revealed that a subset of mice exhibited conspicuously high uptake of glucose in the ipsilateral hemisphere at 1 week post-occlusion (asymmetric group), whereas other mice showed symmetric uptake in both hemispheres (symmetric group). Thus, we aimed to understand the discrepancy between the two groups. Cerebral blood flow and histological/metabolic changes were analyzed using laser Doppler flowmetry and immunohistochemistry/Western blotting, respectively. Contrary to the increased glucose uptake observed in the ischemic cerebral hemisphere on FDG PET (p<0.001), cerebral blood flow tended to be lower in the asymmetric group than in the symmetric group (right to left ratio [%], 36.4±21.8 vs. 58.0±24.8, p=0.059). Neuronal death was observed only in the ischemic hemisphere of the asymmetric group. In contrast, astrocytes were more activated in the asymmetric group than in the symmetric group (p<0.05). Glucose transporter-1, and monocarboxylate transporter-1 were also upregulated in the asymmetric group, compared with the symmetric group (p<0.05, respectively). These results suggest that the increased FDG uptake was associated with relatively severe ischemia, and glucose transporter-1 upregulation and astrocyte activation. Glucose metabolism may thus be a compensatory mechanism in the moderately severe ischemic brain.
Collapse
Affiliation(s)
- Jin Soo Lee
- Department of Neurology, Ajou University School of Medicine, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| | - Ji Man Hong
- Department of Neurology, Ajou University School of Medicine, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| | - Bok Seon Yoon
- Department of Neurology, Ajou University School of Medicine, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| | - Keoung Sun Son
- Department of Neurology, Ajou University School of Medicine, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| | - Kyung Eon Lee
- School of Pharmacy, Kyung Hee University, Seoul 02447, Korea
| | - Doo Soon Im
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Bok-Nam Park
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Korea
| | - Young-Sil An
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Korea
| | - Dong Hoon Hwang
- Department of Brain Science, Ajou University School of Medicine, Korea
| | - Chan Bae Park
- Department of Biology, Ajou University School of Medicine, Korea
| | - Byung Gon Kim
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Korea.,Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
18
|
Marushima A, Nieminen M, Kremenetskaia I, Gianni-Barrera R, Woitzik J, von Degenfeld G, Banfi A, Vajkoczy P, Hecht N. Balanced single-vector co-delivery of VEGF/PDGF-BB improves functional collateralization in chronic cerebral ischemia. J Cereb Blood Flow Metab 2020; 40:404-419. [PMID: 30621518 PMCID: PMC7370608 DOI: 10.1177/0271678x18818298] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The myoblast-mediated delivery of angiogenic genes represents a cell-based approach for targeted induction of therapeutic collateralization. Here, we tested the superiority of myoblast-mediated co-delivery of vascular endothelial growth factor-A (VEGF) together with platelet-derived growth factor-BB (PDGF-BB) on transpial collateralization of an indirect encephalomyosynangiosis (EMS) in a model of chronic cerebral ischemia. Mouse myoblasts expressing a reporter gene alone (empty vector), VEGF, PDGF-BB or VEGF and PDGF-BB through a single bi-cistronic vector (VIP) were implanted into the temporalis muscle of an EMS following permanent ipsilateral internal carotid artery occlusion in adult, male C57BL/6N mice. Over 84 days, myoblast engraftment and gene product expression, hemodynamic impairment, transpial collateralization, angiogenesis, pericyte recruitment and post-ischemic neuroprotection were assessed. By day 42, animals that received PDGF-BB in combination with VEGF (VIP) showed superior hemodynamic recovery, EMS collateralization and ischemic protection with improved pericyte recruitment around the parenchymal vessels and EMS collaterals. Also, supplementation of PDGF-BB resulted in a striking astrocytic activation with intrinsic VEGF mobilization in the cortex below the EMS. Our findings suggest that EMS surgery together with myoblast-mediated co-delivery of VEGF/PDGF-BB may have the potential to serve as a novel treatment strategy for augmentation of collateral flow in the chronically hypoperfused brain.
Collapse
Affiliation(s)
- Aiki Marushima
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Melina Nieminen
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roberto Gianni-Barrera
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Johannes Woitzik
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Andrea Banfi
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Peter Vajkoczy
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Hecht
- Department of Neurosurgery and Center for Stroke research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
19
|
Effects of Tai Chi on Cerebral Hemodynamics and Health-Related Outcomes in Older Community Adults at Risk of Ischemic Stroke: A Randomized Controlled Trial. J Aging Phys Act 2019; 27:678–687. [DOI: 10.1123/japa.2018-0232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study investigated the effects of Tai Chi compared with no exercise control on the cerebral hemodynamic parameters and other health-related factors in community older adults at risk of ischemic stroke. A total of 170 eligible participants were randomly allocated to Tai Chi or control group. The cerebral hemodynamic parameters and physical fitness risk factors of cardiovascular disease were measured at baseline, 12 weeks, and 24 weeks. After the 12-week intervention, Tai Chi significantly improved the minimum of blood flow velocity (BFVmin); BFVmean; pulsatility index and resistance index of the right anterior cerebral artery; and BFVmax, BFVmin, and BFVmeanparameters of the right middle cerebral artery. Tai Chi training also decreased triglyceride, fasting blood glucose, and homocysteine levels, and improved balance ability. Therefore, the supervised 12-week Tai Chi exercise had potential beneficial effects on cerebral hemodynamics, plasma risk factors, and balance ability in older community adults at risk of ischemic stroke.
Collapse
|
20
|
Collateral Vessels Have Unique Endothelial and Smooth Muscle Cell Phenotypes. Int J Mol Sci 2019; 20:ijms20153608. [PMID: 31344780 PMCID: PMC6695737 DOI: 10.3390/ijms20153608] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Collaterals are unique blood vessels present in the microcirculation of most tissues that, by cross-connecting a small fraction of the outer branches of adjacent arterial trees, provide alternate routes of perfusion. However, collaterals are especially susceptible to rarefaction caused by aging, other vascular risk factors, and mouse models of Alzheimer’s disease—a vulnerability attributed to the disturbed hemodynamic environment in the watershed regions where they reside. We examined the hypothesis that endothelial and smooth muscle cells (ECs and SMCs, respectively) of collaterals have specializations, distinct from those of similarly-sized nearby distal-most arterioles (DMAs) that maintain collateral integrity despite their continuous exposure to low and oscillatory/disturbed shear stress, high wall stress, and low blood oxygen. Examination of mouse brain revealed the following: Unlike the pro-inflammatory cobble-stoned morphology of ECs exposed to low/oscillatory shear stress elsewhere in the vasculature, collateral ECs are aligned with the vessel axis. Primary cilia, which sense shear stress, are present, unexpectedly, on ECs of collaterals and DMAs but are less abundant on collaterals. Unlike DMAs, collaterals are continuously invested with SMCs, have increased expression of Pycard, Ki67, Pdgfb, Angpt2, Dll4, Ephrinb2, and eNOS, and maintain expression of Klf2/4. Collaterals lack tortuosity when first formed during development, but tortuosity becomes evident within days after birth, progresses through middle age, and then declines—results consistent with the concept that collateral wall cells have a higher turnover rate than DMAs that favors proliferative senescence and collateral rarefaction. In conclusion, endothelial and SMCs of collaterals have morphologic and functional differences from those of nearby similarly sized arterioles. Future studies are required to determine if they represent specializations that counterbalance the disturbed hemodynamic, pro-inflammatory, and pro-proliferative environment in which collaterals reside and thus mitigate their risk factor-induced rarefaction.
Collapse
|
21
|
Genetic and Environmental Contributions to Variation in the Posterior Communicating Collaterals of the Circle of Willis. Transl Stroke Res 2019; 10:189-203. [PMID: 29589286 DOI: 10.1007/s12975-018-0626-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 02/07/2023]
Abstract
Variation in blood flow mediated by the posterior communicating collateral arteries (PComs) contributes to variation in the severity of tissue injury in obstructive disease. Evidence in animals and humans indicates that differences in the extent of PComs, i.e., their anatomic lumen diameter and whether they are present bilaterally, unilaterally, or absent, are a major factor. These differences arise during development since they are present at birth. However, the causal mechanisms are unknown. We used angiography after maximal dilation to examine involvement of genetic, environmental, and stochastic factors. The extent of PComs varied widely among seven genetically diverse strains of mice. Like pial collaterals in the microcirculation, aging and hypertension reduced PCom diameter, while in contrast, obesity, hyperlipidemia, metabolic syndrome, and diabetes mellitus had no effect. Naturally occurring intrauterine growth restriction had no effect on extent of PCom or pial collaterals in the adult. The number and diameter of PComs evidenced much larger apparent stochastic-dependent variation than pial collaterals. In addition, both PComs underwent flow-mediated outward remodeling after unilateral permanent MCA occlusion that varied with genetic background and was greater on the ipsilesional side. These findings indicate that variation in the number and diameter of PCom collateral arteries arises from stochastic factors and naturally occurring genetic variants that differ from those that cause variation in pial collateral arterioles. Environmental factors also contribute: aging and hypertension reduce PCom diameter. Our results suggest possible sources of variation of PComs in humans and provide information relevant when studying mouse models of occlusive cerebrovascular disease.
Collapse
|
22
|
Rzechorzek W, Zhang H, Buckley BK, Hua K, Pomp D, Faber JE. Aerobic exercise prevents rarefaction of pial collaterals and increased stroke severity that occur with aging. J Cereb Blood Flow Metab 2017; 37:3544-3555. [PMID: 28685617 PMCID: PMC5669350 DOI: 10.1177/0271678x17718966] [Citation(s) in RCA: 36] [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: 01/05/2017] [Revised: 06/07/2017] [Accepted: 06/10/2017] [Indexed: 11/16/2022]
Abstract
Variation in extent of the brain's collateral circulation is an important determinant of variation in the severity of stroke and efficacy of revascularization therapies. However, the number and diameter of pial collateral "arterioles" decrease with aging in associated with reduced eNOS and increased oxidative stress. We tested whether exercise reduces this aging-induced rarefaction. Twelve-month-old mice were randomized to sedentary or voluntary wheel-running. At 26 months' age, permanent MCA occlusion was followed 72 h later by determination of infarct volume and vascular casting after maximal dilation. The decline in collateral number and diameter and 2.4-fold increase in infarct volume evident in 26-versus 3-month-old sedentary mice were prevented by exercise-training. In contrast, number and diameter of the posterior communicating collateral "arteries" were unaffected by aging or exercise. Interestingly, diameter of the primary intracranial arteries increased with aging. Mechanistically, genetic overexpression of eNOS inhibited age-induced collateral rarefaction, and exercise increased eNOS and SOD2 and decreased the inflammatory marker NFkB assessed in hindlimb arteries. In conclusion, exercise prevented age-induced rarefaction of pial collaterals and reduced infarct volume. Aging also promoted outward remodeling of intracranial arteries. These effects were associated with increased eNOS and reduced markers of inflammation and aging in the vascular wall.
Collapse
Affiliation(s)
- Wojciech Rzechorzek
- Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA
| | - Hua Zhang
- Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA
| | - Brian K Buckley
- Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA
| | - Kunjie Hua
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | - Daniel Pomp
- Department of Genetics, University of North Carolina at Chapel Hill, NC, USA
| | - James E Faber
- Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, NC, USA
- Curriculum in Neurobiology, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
23
|
Matin N, Fisher C, Jackson WF, Diaz-Otero JM, Dorrance AM. Carotid artery stenosis in hypertensive rats impairs dilatory pathways in parenchymal arterioles. Am J Physiol Heart Circ Physiol 2017; 314:H122-H130. [PMID: 28842441 DOI: 10.1152/ajpheart.00638.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hypertension is a leading risk factor for vascular cognitive impairment and is strongly associated with carotid artery stenosis. In normotensive rats, chronic cerebral hypoperfusion induced by bilateral common carotid artery stenosis (BCAS) leads to cognitive impairment that is associated with impaired endothelium-dependent dilation in parenchymal arterioles (PAs). The aim of this study was to assess the effects of BCAS on PA function and structure in stroke-prone spontaneously hypertensive rats, a model of human essential hypertension. Understanding the effects of hypoperfusion on PAs in a hypertensive model could lead to the identification of therapeutic targets for cognitive decline in a model that reflects the at-risk population. We hypothesized that BCAS would impair endothelium-dependent dilation in PAs and induce artery remodeling compared with sham rats. PAs from BCAS rats had endothelial dysfunction, as assessed using pressure myography. Inhibition of nitric oxide and prostaglandin production had no effect on PA dilation in sham or BCAS rats. Surprisingly, inhibition of epoxyeicosatrienoic acid production increased dilation in PAs from BCAS rats but not from sham rats. Similar results were observed in the presence of inhibitors for all three dilatory pathways, suggesting that epoxygenase inhibition may have restored a nitric oxide/prostaglandin-independent dilatory pathway in PAs from BCAS rats. PAs from BCAS rats underwent remodeling with a reduced wall thickness. These data suggest that marked endothelial dysfunction in PAs from stroke-prone spontaneously hypertensive rats with BCAS may be associated with the development of vascular cognitive impairment. NEW & NOTEWORTHY The present study assessed the structure and function of parenchymal arterioles in a model of chronic cerebral hypoperfusion and hypertension, both of which are risk factors for cognitive impairment. We observed that impaired dilation and artery remodeling in parenchymal arterioles and abolished cerebrovascular reserve capacity may mediate cognitive deficits.
Collapse
Affiliation(s)
- Nusrat Matin
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| | - Courtney Fisher
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| | - Janice M Diaz-Otero
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| |
Collapse
|
24
|
Michalski D, Hofmann S, Pitsch R, Grosche J, Härtig W. Neurovascular Specifications in the Alzheimer-Like Brain of Mice Affected by Focal Cerebral Ischemia: Implications for Future Therapies. J Alzheimers Dis 2017; 59:655-674. [DOI: 10.3233/jad-170185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Sarah Hofmann
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Roman Pitsch
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | | | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
25
|
Seaman SA, Cao Y, Campbell CA, Peirce SM. Arteriogenesis in murine adipose tissue is contingent on CD68 + /CD206 + macrophages. Microcirculation 2017; 24:10.1111/micc.12341. [PMID: 27976451 PMCID: PMC5432396 DOI: 10.1111/micc.12341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/05/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The surgical transfer of skin, fat, and/or muscle from a donor site to a recipient site within the same patient is a widely performed procedure in reconstructive surgeries. A surgical pretreatment strategy that is intended to increase perfusion in the flap, termed "flap delay," is a commonly employed technique by plastic surgeons prior to flap transplantation. Here, we explored whether CD68+ /CD206+ macrophages are required for arteriogenesis within the flap by performing gain-of-function and loss-of-function studies in a previously published flap delay murine model. METHODS AND RESULTS Local injection of M2-polarized macrophages into the flap resulted in an increase in collateral vessel diameter. Application of a thin biomaterial film loaded with a pharmacological agent (FTY720), which has been previously shown to recruit CD68+ /CD206+ macrophages to remodeling tissue, increased CD68+ /CD206+ cell recruitment and collateral vessel enlargement. Conversely, when local macrophage populations were depleted within the inguinal fat pad via clodronate liposome delivery, we observed fewer CD68+ cells accompanied by diminished collateral vessel enlargement. CONCLUSIONS Our study underscores the importance of macrophages during microvascular adaptations that are induced by flap delay. These studies suggest a mechanism for a translatable therapeutic target that may be used to enhance the clinical flap delay procedure.
Collapse
Affiliation(s)
- Scott A. Seaman
- Department of Biomedical Engineering, University of Virginia
| | - Yiqi Cao
- Department of Biomedical Engineering, University of Virginia
| | | | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia
- Department of Plastic Surgery, University of Virginia
| |
Collapse
|
26
|
Struys T, Govaerts K, Oosterlinck W, Casteels C, Bronckaers A, Koole M, Van Laere K, Herijgers P, Lambrichts I, Himmelreich U, Dresselaers T. In vivo evidence for long-term vascular remodeling resulting from chronic cerebral hypoperfusion in mice. J Cereb Blood Flow Metab 2017; 37:726-739. [PMID: 26994041 PMCID: PMC5381461 DOI: 10.1177/0271678x16638349] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We have characterized both acute and long-term vascular and metabolic effects of unilateral common carotid artery occlusion in mice by in vivo magnetic resonance imaging and positron emission tomography. This common carotid artery occlusion model induces chronic cerebral hypoperfusion and is therefore relevant to both preclinical stroke studies, where it serves as a control condition for a commonly used mouse model of ischemic stroke, and neurodegeneration, as chronic hypoperfusion is causative to cognitive decline. By using perfusion magnetic resonance imaging, we demonstrate that under isoflurane anesthesia, cerebral perfusion levels recover gradually over one month. This recovery is paralleled by an increase in lumen diameter and altered tortuosity of the contralateral internal carotid artery at one year post-ligation as derived from magnetic resonance angiography data. Under urethane/α-chloralose anesthesia, no acute perfusion differences are observed, but the vascular response capacity to hypercapnia is found to be compromised. These hemispheric perfusion alterations are confirmed by water [15O]-H2O positron emission tomography. Glucose metabolism ([18F]-FDG positron emission tomography) or white matter organization (diffusion-weighted magnetic resonance imaging) did not show any significant alterations. In conclusion, permanent unilateral common carotid artery occlusion results in acute and long-term vascular remodeling, which may have immediate consequences for animal models of stroke but also vascular dementia.
Collapse
Affiliation(s)
- Tom Struys
- 1 Biomedical Research Institute - Morphology Research Group, Hasselt University, Hasselt, Belgium
| | - Kristof Govaerts
- 2 Biomedical MRI Unit - MoSAIC, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Wouter Oosterlinck
- 3 Research Unit of Experimental Cardiac Surgery, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Cindy Casteels
- 4 Nuclear Medicine - MoSAIC, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Annelies Bronckaers
- 1 Biomedical Research Institute - Morphology Research Group, Hasselt University, Hasselt, Belgium
| | - Michel Koole
- 4 Nuclear Medicine - MoSAIC, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Koen Van Laere
- 4 Nuclear Medicine - MoSAIC, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Paul Herijgers
- 3 Research Unit of Experimental Cardiac Surgery, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Ivo Lambrichts
- 1 Biomedical Research Institute - Morphology Research Group, Hasselt University, Hasselt, Belgium
| | - Uwe Himmelreich
- 2 Biomedical MRI Unit - MoSAIC, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium
| | - Tom Dresselaers
- 2 Biomedical MRI Unit - MoSAIC, Department of Imaging & Pathology, KU Leuven, Leuven, Belgium.,5 Radiology, University Hospitals, Leuven, Belgium
| |
Collapse
|
27
|
Faber JE, Moore SM, Lucitti JL, Aghajanian A, Zhang H. Sex Differences in the Cerebral Collateral Circulation. Transl Stroke Res 2016; 8:273-283. [PMID: 27844273 DOI: 10.1007/s12975-016-0508-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/26/2016] [Accepted: 11/01/2016] [Indexed: 12/15/2022]
Abstract
Premenopausal women and intact female rodents sustain smaller cerebral infarctions than males. Several sex-dependent differences have been identified as potential contributors, but many questions remain unanswered. Mice exhibit wide variation in native collateral number and diameter (collateral extent) that is dependent on differences in genetic background, aging, and other comorbidities and that contributes to their also-wide differences in infarct volume. Likewise, variation in infarct volume correlates with differences in collateral-dependent blood flow in patients with acute ischemic stroke. We examined whether extent of pial collateral arterioles and posterior communicating collateral arteries (PComAs) differ depending on sex in young, aged, obese, hypertensive, and genetically different mice. We combined new data with meta-analysis of our previously published data. Females of C57BL/6J (B6) and BALB/cByJ (BC) strains sustained smaller infarctions than males after permanent MCA occlusion. This protection was unchanged in BC mice after introgression of the B6 allele of Dce1, the major genetic determinant of variation in pial collaterals among mouse strains. Consistent with this, collateral extent in these and other strains did not differ with sex. Extent of PComAs and primary cerebral arteries also did not vary with sex. No dimorphism was evident for loss of pial collateral number and/or diameter (collateral rarefaction) caused by aging, obesity, and hypertension, nor for collateral remodeling after pMCAO. However, rarefaction was greater in females with long-standing hypertension. We conclude that smaller infarct volume in female mice is not due to greater collateral extent, greater remodeling, or less rarefaction caused by aging, obesity, or hypertension.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Scott M Moore
- Department of Surgery, University of Colorado, Denver, CO, USA
| | - Jennifer L Lucitti
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Amir Aghajanian
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, The McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
28
|
Seaman SA, Cao Y, Campbell CA, Peirce SM. Macrophage Recruitment and Polarization During Collateral Vessel Remodeling in Murine Adipose Tissue. Microcirculation 2016; 23:75-87. [PMID: 26638986 DOI: 10.1111/micc.12261] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/25/2015] [Indexed: 12/29/2022]
Abstract
OBJECTIVE During autologous flap transplantation for reconstructive surgeries, plastic surgeons use a surgical pre-treatment strategy called "flap delay," which entails ligating a feeding artery into an adipose tissue flap 10-14 days prior to transfer. It is believed that this blood flow alteration leads to vascular remodeling in the flap, resulting in better flap survival following transfer; however, the structural changes in the microvascular network are poorly understood. Here, we evaluate microvascular adaptations within adipose tissue in a murine model of flap delay. METHODS AND RESULTS We used a murine flap delay model in which we ligated an artery supplying the inguinal fat pad. Although the extent of angiogenesis appeared minimal, significant diameter expansion of pre-existing collateral arterioles was observed. There was a 5-fold increase in recruitment of CX3CR1(+) monocytes to ligated tissue, a threefold increase in CD68(+) /CD206(+) macrophages in ligated tissue, a 40% increase in collateral vessel diameters supplying ligated tissue, and a 6-fold increase in the number of proliferating cells in ligated tissue. CONCLUSIONS Our study describes microvascular adaptations in adipose in response to altered blood flow and underscores the importance of macrophages. Our data supports the development of therapies that target macrophages in order to enhance vascular remodeling in flaps.
Collapse
Affiliation(s)
- Scott A Seaman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Yiqi Cao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Chris A Campbell
- Department of Plastic Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Department of Plastic Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
29
|
Hayakawa EH, Matsuoka H. Detailed methodology for high resolution scanning electron microscopy (SEM) of murine malaria parasitized-erythrocytes. Parasitol Int 2016; 65:539-544. [DOI: 10.1016/j.parint.2016.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/01/2016] [Accepted: 03/13/2016] [Indexed: 10/22/2022]
|
30
|
Härtig W, Appel S, Suttkus A, Grosche J, Michalski D. Abolished perineuronal nets and altered parvalbumin-immunoreactivity in the nucleus reticularis thalami of wildtype and 3xTg mice after experimental stroke. Neuroscience 2016; 337:66-87. [PMID: 27634771 DOI: 10.1016/j.neuroscience.2016.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/18/2016] [Accepted: 09/03/2016] [Indexed: 12/14/2022]
Abstract
Treatment strategies for ischemic stroke are still limited, since numerous attempts were successful only in preclinical research but failed under clinical condition. To overcome this translational roadblock, clinical relevant stroke models should consider co-morbidities, age-related effects and the complex neurovascular unit (NVU) concept. The NVU includes neurons, vessels and glial cells with astrocytic endfeet in close relation to the extracellular matrix (ECM). However, the role of the ECM after stroke-related tissue damage is poorly understood and mostly neglected for treatment strategies. This study is focused on alterations of perineuronal nets (PNs) as ECM constituents and parvalbumin-containing GABAergic neurons in mice with emphasis on the nucleus reticularis thalami (NRT) in close proximity to the ischemic lesion as induced by a filament-based stroke model. One day after ischemia onset, immunofluorescence-based quantitative analyses revealed drastically declined PNs in the ischemia-affected NRT from 3- and 12-month-old wildtype and co-morbid triple-transgenic (3xTg) mice with Alzheimer-like alterations. Parvalbumin-positive cells decreased numerically in the ischemia-affected NRT, while staining intensity did not differ between the affected and non-affected hemisphere. Additional qualitative analyses demonstrated ischemia-induced loss of PNs and allocated neuropil ECM immunoreactive for aggrecan and neurocan, and impaired immunoreactivity for calbindin, the potassium channel subunit Kv3.1b and the glutamate decarboxylase isoforms GAD65 and GAD67 in the NRT. In conclusion, these data confirm PNs as highly sensitive constituents of the ECM along with impaired neuronal integrity of GABAergic neurons. Therefore, specific targeting of ECM components might appear as a promising strategy for future treatment strategies in stroke.
Collapse
Affiliation(s)
- Wolfgang Härtig
- Paul Flechsig Institute for Brain Research University of Leipzig, Liebigstr. 19, 04103 Leipzig, Germany.
| | - Simon Appel
- Paul Flechsig Institute for Brain Research University of Leipzig, Liebigstr. 19, 04103 Leipzig, Germany
| | - Anne Suttkus
- Paul Flechsig Institute for Brain Research University of Leipzig, Liebigstr. 19, 04103 Leipzig, Germany; Department of Pediatric Surgery, University Hospital Leipzig, Liebigstr. 20 A, 04103 Leipzig, Germany
| | - Jens Grosche
- Effigos GmbH, Am Deutschen Platz 4, 04103 Leipzig, Germany
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103 Leipzig, Germany
| |
Collapse
|
31
|
Wang XM, Xiao H, Liu LL, Cheng D, Li XJ, Si LY. FGF21 represses cerebrovascular aging via improving mitochondrial biogenesis and inhibiting p53 signaling pathway in an AMPK-dependent manner. Exp Cell Res 2016; 346:147-56. [PMID: 27364911 DOI: 10.1016/j.yexcr.2016.06.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/02/2016] [Accepted: 06/25/2016] [Indexed: 12/21/2022]
Abstract
Cerebrovascular aging has a high relationship with stroke and neurodegenerative disease. In the present study, we evaluated the influence of fibroblast growth factor 21 (FGF21) on angiotensin (Ang II)-mediated cerebrovascular aging in human brain vascular smooth muscle cells (hBVSMCs). Ang II induced remarkable aging-phenotypes in hBVSMCs, including enhanced SA-β-gal staining and NBS1 protein expression. First, we used immunoblotting assay to confirm protein expression of FGF21 receptor (FGFR1) and the co-receptor β-Klotho in cultured hBVSMCs. Second, we found that FGF21 treatment partly prevented the aging-related changes induced by Ang II. FGF21 inhibited Ang II-enhanced ROS production/superoxide anion levels, rescued the Ang II-reduced Complex IV and citrate synthase activities, and suppressed the Ang II-induced meprin protein expression. Third, we showed that FGF21 not only inhibited the Ang II-induced p53 activation, but also blocked the action of Ang II on Siah-1-TRF signaling pathway which is upstream factors for p53 activation. At last, either chemical inhibition of AMPK signaling pathway by a specific antagonist Compound C or knockdown of AMPKα1/2 isoform using siRNA, successfully abolished the anti-aging action of FGF21 in hBVSMCs. These results indicate that FGF21 protects against Ang II-induced cerebrovascular aging via improving mitochondrial biogenesis and inhibiting p53 activation in an AMPK-dependent manner, and highlight the therapeutic value of FGF21 in cerebrovascular aging-related diseases such as stroke and neurodegenerative disease.
Collapse
Affiliation(s)
- Xiao-Mei Wang
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hang Xiao
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ling-Lin Liu
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Dang Cheng
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xue-Jun Li
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Liang-Yi Si
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
32
|
Uekawa K, Hasegawa Y, Senju S, Nakagata N, Ma M, Nakagawa T, Koibuchi N, Kim-Mitsuyama S. Intracerebroventricular Infusion of Angiotensin-(1–7) Ameliorates Cognitive Impairment and Memory Dysfunction in a Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2016; 53:127-33. [DOI: 10.3233/jad-150642] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Ken Uekawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Chuo-ku, Kumamoto, Japan
| | - Yu Hasegawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Chuo-ku, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Chuo-ku, Kumamoto, Japan
| | - Mingjie Ma
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Chuo-ku, Kumamoto, Japan
| | - Takashi Nakagawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Chuo-ku, Kumamoto, Japan
| | - Nobutaka Koibuchi
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Chuo-ku, Kumamoto, Japan
| | - Shokei Kim-Mitsuyama
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Chuo-ku, Kumamoto, Japan
| |
Collapse
|
33
|
Hecht N, Müller MM, Sandow N, Pinczolits A, Vajkoczy P, Woitzik J. Infarct prediction by intraoperative laser speckle imaging in patients with malignant hemispheric stroke. J Cereb Blood Flow Metab 2016; 36:1022-32. [PMID: 26661215 PMCID: PMC4908625 DOI: 10.1177/0271678x15612487] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022]
Abstract
Currently, a reliable method for real-time prediction of ischemia in the human brain is not available. Here, we took a first step towards validating non-invasive intraoperative laser speckle imaging (iLSI) for prediction of infarction in 22 patients undergoing decompressive surgery for treatment of malignant hemispheric stroke. During surgery, cortical perfusion was visualized and recorded in real-time with iLSI. The true morphological infarct extension within the iLSI imaging field was superimposed onto the iLSI blood flow maps according to a postoperative MRI (16 h [95% CI: 13, 19] after surgery) with three-dimensional magnetization-prepared rapid gradient-echo and diffusion-weighted imaging reconstruction. Based on the frequency distribution of iLSI perfusion values within the infarcted and non-infarcted territories, probability curves and perfusion thresholds of normalized cerebral blood flow predictive of eventual infarction or non-infarction were calculated. Intraoperative LSI predicted and excluded cortical ischemia with 95% probability at normalized perfusion levels below 40% and above 110%, respectively, which represented 73% of the entire cortical surface area. Together, our results suggest that iLSI is valid for (pseudo-) quantitative assessment of blood flow in the human brain and may be used to identify tissue at risk for infarction at a given time-point in the course of ischemic stroke.
Collapse
Affiliation(s)
- Nils Hecht
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marc-Michael Müller
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nora Sandow
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandra Pinczolits
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
34
|
A new method for evaluating regional cerebral blood flow changes: Laser speckle contrast imaging in a C57BL/6J mouse model of photothrombotic ischemia. ACTA ACUST UNITED AC 2016; 36:174-180. [PMID: 27072958 DOI: 10.1007/s11596-016-1562-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 12/23/2015] [Indexed: 10/22/2022]
Abstract
The present study aimed to improve the processing of data acquired from laser speckle contrast imaging (LSCI) to provide a standardization method to explore changes in regional cerebral blood flow (rCBF) and to determine the correlations among rCBF, cerebral ischemic lesion volume and microvascular density over time in a focal ischemic region. C57BL/6J mice were subjected to focal photothrombotic (PT) ischemia. rCBF was measured using LSCI at different time points before and after PT ischemia through an intact skull. Standardized rCBF (SrCBF), defined as the ratio of rCBF measured in the ipsilateral region of interest (ROI) to that in the corresponding contralateral region, was calculated to evaluate potential changes. In addition, the volume of the ischemic lesion and the microvascular density were determined using Nissl staining and immunofluorescence, respectively. The relationships among the ischemic lesion volume, microvascular density and SrCBF were analyzed over time. The results showed that the cortical rCBF measured using LSCI following PT ischemia in the C57BL/6J mice gradually increased. Changes in the cerebral ischemic lesion volume were negatively correlated with SrCBF in the ischemic region. Changes in the microvascular density were similar to those observed in SrCBF. Our findings indicate that LSCI is a practical technique for observing changes in murine cortical rCBF without skull opening and for analyzing the relationships among the ischemic lesion volume, microvascular density and SrCBF following focal cerebral ischemia. Preliminary results also suggest that the use of LSCI to observe the formation of collateral circulation is feasible.
Collapse
|
35
|
Ichijo M, Ishibashi S, Li F, Yui D, Miki K, Mizusawa H, Yokota T. Sphingosine-1-Phosphate Receptor-1 Selective Agonist Enhances Collateral Growth and Protects against Subsequent Stroke. PLoS One 2015; 10:e0138029. [PMID: 26367258 PMCID: PMC4569572 DOI: 10.1371/journal.pone.0138029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/24/2015] [Indexed: 11/22/2022] Open
Abstract
Background and Purpose Collateral growth after acute occlusion of an intracranial artery is triggered by increasing shear stress in preexisting collateral pathways. Recently, sphingosine-1-phosphate receptor-1 (S1PR1) on endothelial cells was reported to be essential in sensing fluid shear stress. Here, we evaluated the expression of S1PR1 in the hypoperfused mouse brain and investigated the effect of a selective S1PR1 agonist on leptomeningeal collateral growth and subsequent ischemic damage after focal ischemia. Methods In C57Bl/6 mice (n = 133) subjected to unilateral common carotid occlusion (CCAO) and sham surgery. The first series examined the time course of collateral growth, cell proliferation, and S1PR1 expression in the leptomeningeal arteries after CCAO. The second series examined the relationship between pharmacological regulation of S1PR1 and collateral growth of leptomeningeal anastomoses. Animals were randomly assigned to one of the following groups: LtCCAO and daily intraperitoneal (ip) injection for 7 days of an S1PR1 selective agonist (SEW2871, 5 mg/kg/day); sham surgery and daily ip injection for 7 days of SEW2871 after surgery; LtCCAO and daily ip injection for 7 days of SEW2871 and an S1PR1 inverse agonist (VPC23019, 0.5 mg/kg); LtCCAO and daily ip injection of DMSO for 7 days after surgery; and sham surgery and daily ip injection of DMSO for 7 days. Leptomeningeal anastomoses were visualized 14 days after LtCCAO by latex perfusion method, and a set of animals underwent subsequent permanent middle cerebral artery occlusion (pMCAO) 7days after the treatment termination. Neurological functions 1hour, 1, 4, and 7days and infarction volume 7days after pMCAO were evaluated. Results In parallel with the increase in S1PR1 mRNA levels, S1PR1 expression colocalized with endothelial cell markers in the leptomeningeal arteries, increased markedly on the side of the CCAO, and peaked 7 days after CCAO. Mitotic cell numbers in the leptomeningeal arteries increased after CCAO. Administration of the S1PR1 selective agonist significantly increased cerebral blood flow (CBF) and the diameter of leptomeningeal collateral vessels (42.9 ± 2.6 μm) compared with the controls (27.6 ± 5.7 μm; P < 0.01). S1PR1 inverse agonist administration diminished the effect of the S1PR1 agonist (P < 0.001). After pMCAO, S1PR1 agonist pretreated animals showed significantly smaller infarct volume (17.5% ± 4.0% vs. 7.7% ± 4.0%, P < 0.01) and better functional recovery than vehicle-treated controls. Conclusions These results suggest that S1PR1 is one of the principal regulators of leptomeningeal collateral recruitment at the site of increased shear stress and provide evidence that an S1PR1 selective agonist has a role in promoting collateral growth and preventing of ischemic damage and neurological dysfunction after subsequent stroke in patients with intracranial major artery stenosis or occlusion.
Collapse
Affiliation(s)
- Masahiko Ichijo
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoru Ishibashi
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail:
| | - Fuying Li
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daishi Yui
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazunori Miki
- Department of Endovascular Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidehiro Mizusawa
- Department of Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Tokyo, Japan
- The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
36
|
Starosolski Z, Villamizar CA, Rendon D, Paldino MJ, Milewicz DM, Ghaghada KB, Annapragada AV. Ultra High-Resolution In vivo Computed Tomography Imaging of Mouse Cerebrovasculature Using a Long Circulating Blood Pool Contrast Agent. Sci Rep 2015; 5:10178. [PMID: 25985192 PMCID: PMC4650815 DOI: 10.1038/srep10178] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/01/2015] [Indexed: 12/21/2022] Open
Abstract
Abnormalities in the cerebrovascular system play a central role in many neurologic diseases. The on-going expansion of rodent models of human cerebrovascular diseases and the need to use these models to understand disease progression and treatment has amplified the need for reproducible non-invasive imaging methods for high-resolution visualization of the complete cerebral vasculature. In this study, we present methods for in vivo high-resolution (19 μm isotropic) computed tomography imaging of complete mouse brain vasculature. This technique enabled 3D visualization of large cerebrovascular networks, including the Circle of Willis. Blood vessels as small as 40 μm were clearly delineated. ACTA2 mutations in humans cause cerebrovascular defects, including abnormally straightened arteries and a moyamoya-like arteriopathy characterized by bilateral narrowing of the internal carotid artery and stenosis of many large arteries. In vivo imaging studies performed in a mouse model of Acta2 mutations demonstrated the utility of this method for studying vascular morphometric changes that are practically impossible to identify using current histological methods. Specifically, the technique demonstrated changes in the width of the Circle of Willis, straightening of cerebral arteries and arterial stenoses. We believe the use of imaging methods described here will contribute substantially to the study of rodent cerebrovasculature.
Collapse
Affiliation(s)
- Zbigniew Starosolski
- 1] Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston TX [2] Department of Radiology, Baylor College of Medicine, Houston, TX
| | - Carlos A Villamizar
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - David Rendon
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Michael J Paldino
- 1] Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston TX [2] Department of Radiology, Baylor College of Medicine, Houston, TX
| | - Dianna M Milewicz
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX
| | - Ketan B Ghaghada
- 1] Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston TX [2] Department of Radiology, Baylor College of Medicine, Houston, TX
| | - Ananth V Annapragada
- 1] Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston TX [2] Department of Radiology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
37
|
Impaired leptomeningeal collateral flow contributes to the poor outcome following experimental stroke in the Type 2 diabetic mice. J Neurosci 2015; 35:3851-64. [PMID: 25740515 DOI: 10.1523/jneurosci.3838-14.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Collateral status is an independent predictor of stroke outcome. However, the spatiotemporal manner in which collateral flow maintains cerebral perfusion during cerebral ischemia is poorly understood. Diabetes exacerbates ischemic brain damage, although the impact of diabetes on collateral dynamics remains to be established. Using Doppler optical coherent tomography, a robust recruitment of leptomeningeal collateral flow was detected immediately after middle cerebral artery (MCA) occlusion in C57BL/6 mice, and it continued to grow over the course of 1 week. In contrast, an impairment of collateral recruitment was evident in the Type 2 diabetic db/db mice, which coincided with a worse stroke outcome compared with their normoglycemic counterpart db/+, despite their equally well-collateralized leptomeningeal anastomoses. Similar to the wild-type mice, both db/+ and db/db mice underwent collateral growth 7 d after MCA stroke, although db/db mice still exhibited significantly reduced retrograde flow into the MCA territory chronically. Acutely induced hyperglycemia in the db/+ mice did not impair collateral flow after stroke, suggesting that the state of hyperglycemia alone was not sufficient to impact collateral flow. Human albumin was efficacious in improving collateral flow and outcome after stroke in the db/db mice, enabling perfusion to proximal MCA territory that was usually not reached by retrograde flow from anterior cerebral artery without treatment. Our results suggest that the impaired collateral status contributes to the exacerbated ischemic injury in mice with Type 2 diabetes, and modulation of collateral flow has beneficial effects on stroke outcome among these subjects.
Collapse
|
38
|
Nishijima Y, Akamatsu Y, Weinstein PR, Liu J. Collaterals: Implications in cerebral ischemic diseases and therapeutic interventions. Brain Res 2015; 1623:18-29. [PMID: 25770816 DOI: 10.1016/j.brainres.2015.03.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 02/27/2015] [Accepted: 03/03/2015] [Indexed: 02/09/2023]
Abstract
Despite the tremendous progress made in the treatment of cerebrovascular occlusive diseases, many patients suffering from ischemic brain injury still experience dismal outcomes. Although rehabilitation contributes to post-stroke functional recovery, there is no doubt that interventions that promote the restoration of blood supply are proven to minimize ischemic injury and improve recovery. In response to the acutely decreased blood perfusion during arterial occlusion, arteriogenesis, the compensation of blood flow through the collateral circulation during arterial obstructive diseases can act not only in a timely fashion but also much more efficiently compared to angiogenesis, the sprouting of new capillaries, and a mechanism occurring in a delayed fashion while increases the total resistance of the vascular bed of the affected territory. Interestingly, despite the vast differences between the two vascular remodeling mechanisms, some crucial growth factors and cytokines involved in angiogenesis are also required for arteriogenesis. Understanding the mechanisms underlying vascular remodeling after ischemic brain injury is a critical step towards the development of effective therapies for ischemic stroke. The present article will discuss our current views in vascular remodeling acutely after brain ischemia, namely arteriogenesis, and some relevant clinical therapies available on the horizon in augmenting collateral flow that hold promise in treating ischemic brain injury. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yasuo Nishijima
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA; Department of Neurosurgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yosuke Akamatsu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA; Department of Neurosurgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Phillip R Weinstein
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94121, USA; SFVAMC, San Francisco, CA 94121, USA.
| |
Collapse
|
39
|
Broggini T, Czabanka M, Piffko A, Harms C, Hoffmann C, Mrowka R, Wenke F, Deutsch U, Grötzinger C, Vajkoczy P. ICAM1 depletion reduces spinal metastasis formation in vivo and improves neurological outcome. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2015; 24:2173-81. [PMID: 25711910 DOI: 10.1007/s00586-015-3811-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Clinical treatment of spinal metastasis is gaining in complexity while the underlying biology remains unknown. Insufficient biological understanding is due to a lack of suitable experimental animal models. Intercellular adhesion molecule-1 (ICAM1) has been implicated in metastasis formation. Its role in spinal metastasis remains unclear. It was the aim to generate a reliable spinal metastasis model in mice and to investigate metastasis formation under ICAM1 depletion. MATERIAL AND METHODS B16 melanoma cells were infected with a lentivirus containing firefly luciferase (B16-luc). Stable cell clones (B16-luc) were injected retrogradely into the distal aortic arch. Spinal metastasis formation was monitored using in vivo bioluminescence imaging/MRI. Neurological deficits were monitored daily. In vivo selected, metastasized tumor cells were isolated (mB16-luc) and reinjected intraarterially. mB16-luc cells were injected intraarterially in ICAM1 KO mice. Metastasis distribution was analyzed using organ-specific fluorescence analysis. RESULTS Intraarterial injection of B16-luc and metastatic mB16-luc reliably induced spinal metastasis formation with neurological deficits (B16-luc:26.5, mB16-luc:21 days, p<0.05). In vivo selection increased the metastatic aggressiveness and led to a bone specific homing phenotype. Thus, mB16-luc cells demonstrated higher number (B16-luc: 1.2±0.447, mB16-luc:3.2±1.643) and increased total metastasis volume (B16-luc:2.87±2.453 mm3, mB16-luc:11.19±3.898 mm3, p<0.05) in the spine. ICAM1 depletion leads to a significantly reduced number of spinal metastasis (mB16-luc:1.2±0.84) with improved neurological outcome (29 days). General metastatic burden was significantly reduced under ICAM1 depletion (control: 3.47×10(7)±1.66×10(7); ICAM-1-/-: 5.20×10(4)±4.44×10(4), p<0.05 vs. control) CONCLUSION Applying a reliable animal model for spinal metastasis, ICAM1 depletion reduces spinal metastasis formation due to an organ-unspecific reduction of metastasis development.
Collapse
Affiliation(s)
- Thomas Broggini
- Department of Neurosurgery, Universitätsmedizin Charite, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Universitätsmedizin Charite, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Andras Piffko
- Department of Neurosurgery, Universitätsmedizin Charite, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Universitätsmedizin Charite, Berlin, Germany
| | - Christian Hoffmann
- Department of Experimental Neurology, Universitätsmedizin Charite, Berlin, Germany
| | - Ralf Mrowka
- Experimental Nephrology, Universitätsklinikum, Jena, Germany
| | - Frank Wenke
- Experimental Nephrology, Universitätsklinikum, Jena, Germany
| | - Urban Deutsch
- Theodor Kocher Institute, University of Berne, Berne, Germany
| | - Carsten Grötzinger
- Department for Hepatology and Gastroenterology, Charite, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charite, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
40
|
Leonardo CC, Mendes M, Ahmad AS, Doré S. Efficacy of prophylactic flavan-3-ol in permanent focal ischemia in 12-mo-old mice. Am J Physiol Heart Circ Physiol 2015; 308:H583-91. [PMID: 25576625 DOI: 10.1152/ajpheart.00239.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The consumption of flavan-3-ol-containing foods, including (-)-epicatechin (EC), has been linked to lower incidence of cardiovascular disease and stroke. We previously demonstrated nuclear transcription factor erythroid 2p45-related factor-2 (Nrf2) -dependent EC efficacy in reducing stroke-induced deficits in 2-mo-old mice; yet stroke is primarily a disease of the elderly. Because neuroinflammation, oxidative stress, and vascular dysfunction are hallmarks of aging, we tested whether Nrf2 mediates EC efficacy in aging mice through modulation of glial responses and blood brain barrier permeability. First, we compared anastomosis in naïve wild-type and C57BL/6 Nrf2(-/-) mice to identify potential differences in cerebrovascular architecture. Data showed no significant differences in the number of anastomoses or mean intersection points, indicating similar gross vascular physiology. To assess efficacy and mechanisms of protection, wild-type or Nrf2(-/-) mice were administered the minimum effective EC dose established in our previous studies before the permanent distal middle cerebral artery occlusion. Similar to previous results with young mice, 12-mo-old wild types also showed significant reductions in infarct volume (41.01 ± 29.57%) and improved performance in removing adhesive tape relative to vehicle-treated controls, whereas a trend toward protection was observed in Nrf2(-/-). However, EC did not reduce immunoreactivity for the microglia/macrophage marker anti-ionized calcium-binding adapter molecule 1, suggesting that dampened activation/recruitment did not account for EC protection. Furthermore, there were no differences in mouse IgG extravasation or spontaneous hemorrhage between EC-treated groups. These data demonstrate that EC protection occurs independent of microglia/macrophage modulation or blood brain barrier preservation, suggesting that the glial cell responses in young mice are compensatory to another, and potentially novel, protective mechanism.
Collapse
Affiliation(s)
- Christopher C Leonardo
- Department of Anesthesiology and Center for Translational Research and Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida; and
| | - Monique Mendes
- Department of Anesthesiology and Center for Translational Research and Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida; and
| | - Abdullah S Ahmad
- Department of Anesthesiology and Center for Translational Research and Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida; and
| | - Sylvain Doré
- Department of Anesthesiology and Center for Translational Research and Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, Florida; and Departments of Neurology, Psychiatry and Neuroscience, College of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
41
|
Danière F, Lobotesis K, Machi P, Eker O, Mourand I, Riquelme C, Ayrignac X, Vendrell JF, Gascou G, Fendeleur J, Dargazanli C, Schaub R, Brunel H, Arquizan C, Bonafé A, Costalat V. Patient selection for stroke endovascular therapy--DWI-ASPECTS thresholds should vary among age groups: insights from the RECOST study. AJNR Am J Neuroradiol 2015; 36:32-9. [PMID: 25273535 DOI: 10.3174/ajnr.a4104] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The purpose of this study was to evaluate the benefits of endovascular intervention in large-vessel occlusion strokes, depending on age class. MATERIALS AND METHODS A clinical management protocol including intravenous treatment and mechanical thrombectomy was instigated in our center in 2009 (Prognostic Factors Related to Clinical Outcome Following Thrombectomy in Ischemic Stroke [RECOST] study). All patients with acute ischemic stroke with an anterior circulation major-vessel occlusion who presented within 6 hours were evaluated with an initial MR imaging examination and were analyzed according to age subgroups (younger than 50 years, 50-59 years, 60-69 years, 70-79 years; 80 years or older). The mRS score at 3 months was the study end point. RESULTS One hundred sixty-five patients were included in the analysis. The mean age was 67.4 years (range, 29-90 years). The mean baseline NIHSS score was 17.24 (range, 3-27). The mean DWI-derived ASPECTS was 6.4. Recanalization of TICI 2b/3 was achieved in 80%. At 3 months, 41.72% of patients had a good outcome, with a gradation of prognosis depending on the age subgroup and a clear cutoff at 70 years. Only 19% of patients older than 80 years had a good outcome at 3 months (mean ASPECTS = 7.4) with 28% for 70-79 years (mean ASPECTS = 6.8), but 58% for 60-69 years (mean ASPECTS = 6), 52% for 50-59 years (mean ASPECTS = 5.91), and 72% for younger than 50 years (mean ASPECTS = 6.31). In contrast, the mortality rate was 35% for 80 years and older, and 26% for 70-79 versus 5%-9% for younger than 70 years. CONCLUSIONS The elderly may benefit from thrombectomy when their ischemic core volume is low in comparison with younger patients who still benefit from acute recanalization despite larger infarcts. Stroke volume thresholds should, therefore, be related and adjusted to the patient's age group.
Collapse
Affiliation(s)
- F Danière
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | - K Lobotesis
- Imaging Department (K.L.), Imperial College Healthcare National Health Service Trust, Charing Cross Hospital, London, United Kingdom
| | - P Machi
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | - O Eker
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | | | - C Riquelme
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | | | - J F Vendrell
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | - G Gascou
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | - J Fendeleur
- Anesthesiology (J.F.), CHU Montpellier, Montpellier, France
| | - C Dargazanli
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | - R Schaub
- Department of Medical Statistics (R.S.), CHU Montpellier, Arnaud de Villeneuve Hospital, University of Montpellier, Montpellier, France
| | - H Brunel
- Department of Neuroradiology (H.B.), CHU Marseille, Hôpital La Timone, Marseille, France
| | | | - A Bonafé
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| | - V Costalat
- From the Departments of Neuroradiology (F.D., P.M., O.E., C.R., J.F.V., G.G., C.D., A.B., V.C.)
| |
Collapse
|
42
|
Hecht N, Marushima A, Nieminen M, Kremenetskaia I, von Degenfeld G, Woitzik J, Vajkoczy P. Myoblast-mediated gene therapy improves functional collateralization in chronic cerebral hypoperfusion. Stroke 2014; 46:203-11. [PMID: 25388423 DOI: 10.1161/strokeaha.114.006712] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Direct extracranial-intracranial bypass surgery for treatment of cerebral hemodynamic compromise remains hindered by complications but alternative simple and safe indirect revascularization procedures, such as an encephalomyosynangiosis (EMS), lack hemodynamic efficiency. Here, the myoblast-mediated transfer of angiogenic genes presents an approach for induction of therapeutic collateralization. In this study, we tested the effect of myoblast-mediated delivery of vascular endothelial growth factor-A (VEGF) to the muscle/brain interface of an EMS in a model of chronic cerebral hypoperfusion. METHODS Permanent unilateral internal carotid artery-occlusion was performed in adult C57/BL6 mice with or without (no EMS) surgical grafting of an EMS followed by implantation of monoclonal mouse myoblasts expressing either VEGF164 or an empty vector (EV). Cerebral hemodynamic impairment, transpial collateralization, angiogenesis, mural cell investment, microvascular permeability, and cortical infarction after ipsilateral stroke were assessed by real-time laser speckle blood flow imaging, 2- and 3-dimensional immunofluorescence and MRI. RESULTS VEGF-expressing myoblasts improved hemodynamic rescue by day 14 (no EMS 37±21%, EV 42±9%, VEGF 48±12%; P<0.05 for VEGF versus no EMS and versus EV), together with the EMS take rate (VEGF 60%, EV 18.2%; P<0.05) and angiogenesis of mature cortical microvessels below the EMS (P<0.05 for VEGF versus EV). Importantly, functional and morphological results were paralleled by a 25% reduction of cortical infarction after experimental stroke on the side of the EMS. CONCLUSIONS Myoblast-mediated VEGF supplementation at the target site of an EMS could help overcome the clinical dilemma of poor surgical revascularization results and provide protection from ischemic stroke.
Collapse
Affiliation(s)
- Nils Hecht
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.)
| | - Aiki Marushima
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.)
| | - Melina Nieminen
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.)
| | - Irina Kremenetskaia
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.)
| | - Georges von Degenfeld
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.)
| | - Johannes Woitzik
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.)
| | - Peter Vajkoczy
- From the Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Germany (N.H., A.M., M.N., I.K., J.W., P.V.); and Baxter Laboratory for Stem Cell Biology, Stanford University, CA (G.v.D.).
| |
Collapse
|
43
|
Collateralization and ischemia in hemodynamic cerebrovascular insufficiency. Acta Neurochir (Wien) 2014; 156:2051-8; discussion 2058. [PMID: 25253629 DOI: 10.1007/s00701-014-2227-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/01/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Moyamoya disease and atherosclerotic cerebrovascular occlusive disease lead to hemodynamic impairment of cerebral blood flow. One major differentiation between both disease entities lies in the collateralization pathways. The clinical implications of the collateralization pathways for the development of hemodynamic ischemia remain unknown. The aim was to characterize collateralization and ischemia patterns in patients with chronic hemodynamic compromise. METHODS Hemodynamic compromise was verified using acetazolamide-stimulated xenon-CT or SPECT in 54 patients [30 moyamoya and 24 atherosclerotic cerebrovascular disease (ACVD)]. All patients received MRI to differentiate hemodynamic ischemia into anterior/posterior cortical border zone infarction (CBI), inferior border zone infarction (IBI) or territorial infarction (TI). Digital subtraction angiography was applied to evaluate collateralization. Collateralization was compared and correlated with the localization of ischemia and number of vascular territories with impaired cerebrovascular reserve capacity (CVRC). RESULTS MM patients showed collateralization significantly more often via pericallosal anastomosis and the posterior communicating artery (flow in the anterior-posterior direction; MM: 95%/95% vs. ACVD: 23%/12%, p < 0.05). ACVD patients demonstrated collateralization via the anterior and posterior communicating arteries (flow in the posterior-anterior direction, MM: 6%/5% vs. ACVD: 62%/88%, p < 0.05). Patterns of infarction were comparable (aCBI: MM: 36% vs. ACVD: 35%; pCBI: MM: 10% vs. ACVD: 20%; IBI: MM: 35% vs. ACVD: 41%; TI: MM: 13% vs. ACVD: 18%). The number and localization of vascular territories with impaired CVRC were comparable. CONCLUSIONS Despite significant differences in collateralization, the infarct patterns and severity of CVRC impairment do not differ between MMV and ACVD patients. Cerebral collateralization does not allow reaching conclusions about the localization of cerebral ischemia or severity of impaired CVRC in chronic hemodynamic impairment.
Collapse
|
44
|
Adamczak JM, Schneider G, Nelles M, Que I, Suidgeest E, van der Weerd L, Löwik C, Hoehn M. In vivo bioluminescence imaging of vascular remodeling after stroke. Front Cell Neurosci 2014; 8:274. [PMID: 25249937 PMCID: PMC4155794 DOI: 10.3389/fncel.2014.00274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/21/2014] [Indexed: 12/14/2022] Open
Abstract
Thrombolysis remains the only beneficial therapy for ischemic stroke, but is restricted to a short therapeutic window following the infarct. Currently research is focusing on spontaneous regenerative processes during the sub-acute and chronic phase. Angiogenesis, the formation of new blood vessels from pre-existing ones, was observed in stroke patients, correlates with longer survival and positively affects the formation of new neurons. Angiogenesis takes place in the border zones of the infarct, but further insight into the temporal profile is needed to fully apprehend its therapeutic potential and its relevance for neurogenesis and functional recovery. Angiogenesis is a multistep process, involving extracellular matrix degradation, endothelial cell proliferation, and, finally, new vessel formation. Interaction between vascular endothelial growth factor and its receptor 2 (VEGFR2) plays a central role in these angiogenic signaling cascades. In the present study we investigated non-invasively the dynamics of VEGFR2 expression following cerebral ischemia in a mouse model of middle cerebral artery occlusion (MCAO). We used a transgenic mouse expressing firefly luciferase under the control of the VEGFR2 promotor to non-invasively elucidate the temporal profile of VEGFR2 expression after stroke as a biomarker for VEGF/VEGFR2 signaling. We measured each animal repetitively up to 2 weeks after stroke and found increased VEGFR2 expression starting 3 days after the insult with peak values at 7 days. These were paralleled by increased VEGFR2 protein levels and increased vascular volume in peri-infarct areas at 14 days after the infarct, indicating that signaling via VEGFR2 leads to successful vascular remodeling. This study describes VEGFR2-related signaling is active at least up to 2 weeks after the infarct and results in increased vascular volume. Further, this study presents a novel strategy for the non-invasive evaluation of angiogenesis-based therapies.
Collapse
Affiliation(s)
- Joanna M Adamczak
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany
| | - Gabriele Schneider
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany
| | - Melanie Nelles
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany
| | - Ivo Que
- Department of Endocrinology, Leiden University Medical Center Leiden, Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center Leiden, Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center Leiden, Netherlands ; Department of Human Genetics, Leiden University Medical Center Leiden, Netherlands
| | - Clemens Löwik
- Department of Endocrinology, Leiden University Medical Center Leiden, Netherlands ; Department of Radiology, Leiden University Medical Center Leiden, Netherlands
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research Cologne, Germany ; Department of Radiology, Leiden University Medical Center Leiden, Netherlands
| |
Collapse
|
45
|
Changes in cortical microvasculature during misery perfusion measured by two-photon laser scanning microscopy. J Cereb Blood Flow Metab 2014; 34:1363-72. [PMID: 24849667 PMCID: PMC4126097 DOI: 10.1038/jcbfm.2014.91] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/02/2014] [Accepted: 04/25/2014] [Indexed: 12/29/2022]
Abstract
This study aimed to examine the cortical microvessel diameter response to hypercapnia in misery perfusion using two-photon laser scanning microscopy (TPLSM). We evaluated whether the vascular response to hypercapnia could represent the cerebrovascular reserve. Cerebral blood flow (CBF) during normocapnia and hypercapnia was measured by laser-Doppler flowmetry through cranial windows in awake C57/BL6 mice before and at 1, 7, 14, and 28 days after unilateral common carotid artery occlusion (UCCAO). Diameters of the cortical microvessels during normocapnia and hypercapnia were also measured by TPLSM. Cerebral blood flow and the vascular response to hypercapnia were decreased after UCCAO. Before UCCAO, vasodilation during hypercapnia was found primarily in arterioles (22.9%±3.5%). At 14 days after UCCAO, arterioles, capillaries, and venules were autoregulatorily dilated by 79.5%±19.7%, 57.2%±32.3%, and 32.0%±10.8%, respectively. At the same time, the diameter response to hypercapnia in arterioles was significantly decreased to 1.9%±1.5%. A significant negative correlation was observed between autoregulatory vasodilation and the diameter response to hypercapnia in arterioles. Our findings indicate that arterioles play main roles in both autoregulatory vasodilation and hypercapnic vasodilation, and that the vascular response to hypercapnia can be used to estimate the cerebrovascular reserve.
Collapse
|
46
|
Hecht N, Schneider UC, Czabanka M, Vinci M, Hatzopoulos AK, Vajkoczy P, Woitzik J. Endothelial progenitor cells augment collateralization and hemodynamic rescue in a model of chronic cerebral ischemia. J Cereb Blood Flow Metab 2014; 34:1297-305. [PMID: 24780900 PMCID: PMC4126089 DOI: 10.1038/jcbfm.2014.78] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/10/2014] [Accepted: 04/04/2014] [Indexed: 01/17/2023]
Abstract
Surgical flow augmentation for treatment of cerebral hemodynamic impairment remains controversial. Here, we investigated the benefit of endothelial progenitor cell (EPC) treatment in a rat model of chronic cerebral hypoperfusion. At repeated time points after 3-vessel occlusion (3-VO), animals were treated with 1 × 10(6) DiI-labeled (a) ex vivo-expanded embryonic-EPC (e-EPC), (b) cyclic AMP-differentiated embryonic-endothelial progenitor-derived cells (e-EPDC as biologic control) or, (c) saline. The cerebrovascular reserve capacity (CVRC) was assessed immediately before and on days 7 and 21 after 3-VO. Structural effects were assessed by latex perfusion, immunohistochemistry, and intravital fluorescence video microscopy on day 21. Three-vessel occlusion resulted in a significant impairment of the CVRC with better functional recovery after treatment with e-EPC (16.4±8%) compared with e-EPDC (3.7±8%) or saline (6.4±9%) by day 21 (P<0.05), which was paralleled by a significant increase in the vessel diameters of the anterior Circle of Willis, a significantly higher number of leptomeningeal anastomoses and higher parenchymal capillary density in e-EPC-treated animals. Interestingly, despite in vivo interaction of e-EPC with the cerebral endothelium, e-EPC incorporation into the cerebral vasculature was not observed. Our results suggest that EPC may serve as a novel therapeutic agent in clinical trials for nonsurgical treatment of chronic cerebral hemodynamic impairment.
Collapse
Affiliation(s)
- Nils Hecht
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ulf C Schneider
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Czabanka
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Vinci
- Department of Neurosurgery, Universitätsmedizin Mannheim, Berlin, Germany
| | - Antonis K Hatzopoulos
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Peter Vajkoczy
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Woitzik
- Department of Neurosurgery and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
47
|
Faber JE, Chilian WM, Deindl E, van Royen N, Simons M. A brief etymology of the collateral circulation. Arterioscler Thromb Vasc Biol 2014; 34:1854-9. [PMID: 25012127 DOI: 10.1161/atvbaha.114.303929] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It is well known that the protective capacity of the collateral circulation falls short in many individuals with ischemic disease of the heart, brain, and lower extremities. In the past 15 years, opportunities created by molecular and genetic tools, together with disappointing outcomes in many angiogenic trials, have led to a significant increase in the number of studies that focus on: understanding the basic biology of the collateral circulation; identifying the mechanisms that limit the collateral circulation's capacity in many individuals; devising methods to measure collateral extent, which has been found to vary widely among individuals; and developing treatments to increase collateral blood flow in obstructive disease. Unfortunately, accompanying this increase in reports has been a proliferation of vague terms used to describe the disposition and behavior of this unique circulation, as well as the increasing misuse of well-ensconced ones by new (and old) students of collateral circulation. With this in mind, we provide a brief glossary of readily understandable terms to denote the formation, adaptive growth, and maladaptive rarefaction of collateral circulation. We also propose terminology for several newly discovered processes that occur in the collateral circulation. Finally, we include terms used to describe vessels that are sometimes confused with collaterals, as well as terms describing processes active in the general arterial-venous circulation when ischemic conditions engage the collateral circulation. We hope this brief review will help unify the terminology used in collateral research.
Collapse
Affiliation(s)
- James E Faber
- From the Departments of Cell Biology and Physiology, University of North Carolina, Chapel Hill (J.E.F.); Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown (W.M.C.), Walter-Brendel-Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany (E.D.); Division of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (N.V.R.); and Departments of Internal Medicine and Cell Biology, Yale Cardiovascular Research Center, New Haven, CT (M.S.).
| | - William M Chilian
- From the Departments of Cell Biology and Physiology, University of North Carolina, Chapel Hill (J.E.F.); Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown (W.M.C.), Walter-Brendel-Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany (E.D.); Division of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (N.V.R.); and Departments of Internal Medicine and Cell Biology, Yale Cardiovascular Research Center, New Haven, CT (M.S.)
| | - Elisabeth Deindl
- From the Departments of Cell Biology and Physiology, University of North Carolina, Chapel Hill (J.E.F.); Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown (W.M.C.), Walter-Brendel-Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany (E.D.); Division of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (N.V.R.); and Departments of Internal Medicine and Cell Biology, Yale Cardiovascular Research Center, New Haven, CT (M.S.)
| | - Niels van Royen
- From the Departments of Cell Biology and Physiology, University of North Carolina, Chapel Hill (J.E.F.); Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown (W.M.C.), Walter-Brendel-Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany (E.D.); Division of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (N.V.R.); and Departments of Internal Medicine and Cell Biology, Yale Cardiovascular Research Center, New Haven, CT (M.S.)
| | - Michael Simons
- From the Departments of Cell Biology and Physiology, University of North Carolina, Chapel Hill (J.E.F.); Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown (W.M.C.), Walter-Brendel-Centre of Experimental Medicine, Ludwig Maximilians University, Munich, Germany (E.D.); Division of Cardiology, VU University Medical Center, Amsterdam, The Netherlands (N.V.R.); and Departments of Internal Medicine and Cell Biology, Yale Cardiovascular Research Center, New Haven, CT (M.S.)
| |
Collapse
|
48
|
Nemeth CL, Gutman DA, Majeed W, Keilholz SD, Neigh GN. Microembolism induces anhedonia but no detectable changes in white matter integrity in aged rats. PLoS One 2014; 9:e96624. [PMID: 24811070 PMCID: PMC4014537 DOI: 10.1371/journal.pone.0096624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 04/09/2014] [Indexed: 11/18/2022] Open
Abstract
Microvascular disease leads to alterations of cerebral vasculature including the formation of microembolic (ME) strokes. Though ME are associated with changes in mood and the severity and progression of cognitive decline, the effect of ME strokes on cerebral microstructure and its relationship to behavioral endpoints is unknown. Here, we used adult and aged male rats to test the hypotheses that ME lesions result in subtle changes to white and gray matter integrity as detected by high-throughput diffusion tensor imaging (DTI) and that these structural disruptions correspond to behavioral deficits. Two weeks post-surgery, aged animals showed depressive-like behaviors in the sucrose consumption test in the absence of altered cerebral diffusivity as assessed by ex-vivo DTI. Furthermore, DTI indices did not correlate with the degree of behavioral disruption in aged animals or in a subset of animals with observed tissue cavitation and subtle DTI alterations. Together, data suggest that behavioral deficits are not the result of damage to brain regions or white matter tracts, rather the activity of other systems may underlie functional disruption and recovery.
Collapse
Affiliation(s)
- Christina L. Nemeth
- Department of Psychiatry and Behavioral Science, Emory University, Atlanta, Georgia, United States of America
- Department of Physiology, Emory University, Atlanta, Georgia, United States of America
| | - David A. Gutman
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia, United States of America
| | - Waqas Majeed
- Coulter Department of Biomedical Engineering, Emory University/Georgia Institute of Technology, Atlanta, Georgia, United States of America
- LUMS, School of Science and Engineering, Department of Electrical Engineering, Lahore, Pakistan
| | - Shella D. Keilholz
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia, United States of America
| | - Gretchen N. Neigh
- Department of Psychiatry and Behavioral Science, Emory University, Atlanta, Georgia, United States of America
- Department of Physiology, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
49
|
Farkas E, Bari F. Spreading depolarization in the ischemic brain: does aging have an impact? J Gerontol A Biol Sci Med Sci 2014; 69:1363-70. [PMID: 24809351 DOI: 10.1093/gerona/glu066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Recurrent waves of spreading depolarization (SD) spontaneously occur minutes after the onset of focal ischemia in the brain and keep generating for a number of days to follow. It has become widely accepted that ischemia-related SDs are part of the pathophysiology of cerebrovascular diseases and predict worse outcome. SDs may exacerbate ischemic injury via related atypical hemodynamic responses. The incidence of ischemic stroke is known to increase markedly with age; yet, very few studies investigated whether age alters SD evolution and whether a potential age-specific pattern of SD would contribute to the age-related intensification of infarct development. Experimental data demonstrate that aging has a marked impact on SD evolution and corresponding changes in cerebral blood flow. We hypothesize that an age-specific pattern of the SD-associated hemodynamic response must be involved in augmenting the expansion of ischemic brain damage in the elderly patients and that structural and functional (mal)adaptation of the cerebrovascular system with aging serves as a potential basis for compromised vascular reactivity and subsequent tissue damage. The concept put forward is expected to stimulate further investigation to achieve a comprehensive overview of the implication of SD in injury progression in the aged brain.
Collapse
Affiliation(s)
- Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| | - Ferenc Bari
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| |
Collapse
|
50
|
Hawkes CA, Michalski D, Anders R, Nissel S, Grosche J, Bechmann I, Carare RO, Härtig W. Stroke-induced opposite and age-dependent changes of vessel-associated markers in co-morbid transgenic mice with Alzheimer-like alterations. Exp Neurol 2013; 250:270-81. [DOI: 10.1016/j.expneurol.2013.09.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 09/17/2013] [Accepted: 09/21/2013] [Indexed: 10/26/2022]
|