1
|
Salim HA, Lakhani DA, Balar AB, Mei J, Luna L, Shahriari M, Hyson NZ, Deng F, Dmytriw AA, Guenego A, Urrutia VC, Marsh EB, Lu H, Xu R, Leigh R, Shah G, Wen S, Albers GW, Hillis AE, Llinas R, Nael K, Wintermark M, Heit JJ, Faizy TD, Yedavalli VS. Factors Associated With Prolonged Venous Transit in Large Vessel Occlusion Acute Ischemic Strokes. J Neuroimaging 2025; 35:e70006. [PMID: 39809719 DOI: 10.1111/jon.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND AND PURPOSE Prolonged venous transit (PVT), derived from computed tomography perfusion (CTP) time-to-maximum (Tmax) maps, reflects compromised venous outflow (VO) in acute ischemic stroke due to large vessel occlusion (AIS-LVO). Poor VO is associated with worse clinical outcomes, but pre-treatment markers predictive of PVT are not well described. METHODS We conducted a retrospective analysis of 189 patients with anterior circulation AIS-LVO who underwent baseline CT evaluation, including non-contrast CT, CT angiography, and CTP. PVT was assessed on Tmax maps; PVT+ was defined as Tmax ≥ 10 s within the posterior superior sagittal sinus or torcula. Baseline clinical data were collected. Multivariable logistic regression identified independent associations between pre-treatment markers and PVT. RESULTS PVT+ was identified in 65 patients (34%). In multivariable analysis, higher admission National Institutes of Health Stroke Scale (NIHSS) scores (adjusted odds ratio [aOR], 1.05 per point; 95% confidence interval [CI], 1.01-1.11; P = 0.028) and male sex (aOR, 1.98; 95% CI, 1.03-3.89; P = 0.043) were independently associated with PVT+. CONCLUSIONS Higher admission NIHSS scores and male sex are independently associated with PVT in anterior circulation AIS-LVO, suggesting that readily available clinical markers may help identify patients with poor VO profiles.
Collapse
Affiliation(s)
- Hamza Adel Salim
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
- Department of Neuroradiology, MD Anderson Medical Center, Houston, Texas, USA
| | - Dhairya A Lakhani
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Aneri B Balar
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Janet Mei
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Licia Luna
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Mona Shahriari
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Nathan Z Hyson
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Francis Deng
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Adam A Dmytriw
- Massachusetts General Hospital, Harvard University, Boston, Massachusetts, USA
- Neurovascular Centre, Departments of Medical Imaging and Neurosurgery, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Adrien Guenego
- Department of Diagnostic and Interventional Neuroradiology, Erasme University Hospital, Brussels, Belgium
| | - Victor C Urrutia
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Elisabeth B Marsh
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Hanzhang Lu
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Risheng Xu
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Rich Leigh
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Gaurang Shah
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sijin Wen
- Department of Epidemiology and Biostatistics, West Virginia University, Morgantown, West Virginia, USA
| | - Gregory W Albers
- Department of Interventional Neuroradiology, Stanford Medical Center, Palo Alto, California, USA
| | - Argye E Hillis
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Rafael Llinas
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| | - Kambiz Nael
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California, USA
| | - Max Wintermark
- Department of Neuroradiology, MD Anderson Medical Center, Houston, Texas, USA
| | - Jeremy J Heit
- Department of Interventional Neuroradiology, Stanford Medical Center, Palo Alto, California, USA
| | - Tobias D Faizy
- Department of Radiology, University Medical Center Münster, Munster, Germany
| | - Vivek S Yedavalli
- Department of Radiology, Division of Neuroradiology, Johns Hopkins Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Haghbin N, Richter DM, Kharche S, Kim MSM, Welsh DG. Functional bias of contractile control in mouse resistance arteries. Sci Rep 2024; 14:24940. [PMID: 39438518 PMCID: PMC11496727 DOI: 10.1038/s41598-024-75838-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Constrictor agonists set arterial tone through two coupling processes, one tied to (electromechanical), the other independent (pharmacomechanical) of, membrane potential (VM). This dual arrangement raises an intriguing question: is the contribution of each mechanism (1) fixed and proportionate, or (2) variable and functionally biased. Examination began in mouse mesenteric arteries with a vasomotor assessment to a classic Gq/11 (phenylephrine) or Gq/11/G12/13 (U46619) agonist, in the absence and presence of nifedipine, to separate among the two coupling mechanisms. Each constrictor elicited a concentration response curve that was attenuated and rightward shifted by nifedipine, findings consistent with functional bias. Electromechanical coupling preceded pharmacomechanical, the latter's importance rising with agonist concentration. In this regard, ensuing contractile and phosphorylation (CPI-17 & MYPT1 (T-855 & T-697)) measures revealed phenylephrine-induced pharmacomechanical coupling was tied to protein kinase C (PKC) activity, while that enabled by U46619 to PKC and Rho-kinase. A complete switch to pharmacomechanical coupling arose when agonist superfusion was replaced by pipet application to a small portion of artery. This switch was predicted, a priori, by a computer model of electromechanical control and supported by additional measures of VM and cytosolic Ca2+. We conclude that the coupling mechanisms driving agonist-induced constriction are variable and functionally biased, their relative importance set in accordance with agonist concentration and manner of application. These findings have important implications to hemodynamic control in health and disease, including hypertension and arterial vasospasm.
Collapse
Affiliation(s)
- Nadia Haghbin
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| | - David M Richter
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Sanjay Kharche
- Department of Medical Biophysics, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Michelle S M Kim
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Donald G Welsh
- Department of Physiology and Pharmacology, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
3
|
Jin X, Imai T, Morais A, Sasaki Y, Chung DY, Ayata C. Hippocampal infarction and generalized seizures predict early mortality after endovascular middle cerebral artery occlusion in mice. Exp Neurol 2024; 380:114903. [PMID: 39079623 PMCID: PMC11347107 DOI: 10.1016/j.expneurol.2024.114903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Endovascular middle cerebral artery occlusion (MCAO) is a widely used experimental ischemic stroke model. However, the model carries high early mortality. Our aim was to investigate the factors that influence early mortality within 48 h of reperfusion after transient MCAO. Using C57BL/6 mice, we induced 1-hour endovascular filament MCAO. To introduce heterogeneity of infarct volumes, a subset of animals had additional tandem common carotid artery occlusion (MCAO+CCAO). Continuous video monitoring was used to gain insight into the cause of death. Mortality within 48 h was 25% in the pooled cohort. All animals with early mortality suffered from infarcts in the hippocampus, sometimes accompanied by infarcts in the thalamus and midbrain, which occurred exclusively in the MCAO+CCAO group. All animals with early mortality developed convulsive seizures captured on video monitoring. None of the animals that did not develop convulsive seizures died. Among the three regions, hippocampal infarction appeared necessary for convulsive seizures and early mortality. Our data highlight seizures as the primary cause of mortality within the first 48 h after endovascular filament MCAO, linked to hippocampal infarction. Since hippocampal blood supply is mainly from the posterior cerebral artery (PCA), avoiding concurrent PCA ischemia can decrease mortality in proximal MCAO models.
Collapse
Affiliation(s)
- Xuyan Jin
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Takahiko Imai
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Andreia Morais
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yuichi Sasaki
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - David Y Chung
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Cenk Ayata
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
4
|
van der Meij A, Holswilder G, Bernsen MLE, van Os HJA, Hofmeijer J, Spaander FHM, Martens JM, van den Wijngaard IR, Lingsma HF, Konduri PR, BLM Majoie C, Schonewille WJ, Dippel DWJ, Kruyt ND, Nederkoorn PJ, van Walderveen MAA, Wermer MJH. Sex differences in clot, vessel and tissue characteristics in patients with a large vessel occlusion treated with endovascular thrombectomy. Eur Stroke J 2024; 9:600-612. [PMID: 38420950 PMCID: PMC11418468 DOI: 10.1177/23969873241231125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION To improve our understanding of the relatively poor outcome after endovascular treatment (EVT) in women we assessed possible sex differences in baseline neuroimaging characteristics of acute ischemic stroke patients with large anterior vessel occlusion (LVO). PATIENTS AND METHODS We included all consecutive patients from the MR CLEAN Registry who underwent EVT between 2014 and 2017. On baseline non-contrast CT and CT angiography, we assessed clot location and clot burden score (CBS), vessel characteristics (presence of atherosclerosis, tortuosity, size, and collateral status), and tissue characteristics with the Alberta Stroke Program Early Computed Tomography Score (ASPECTS). Radiological outcome was assessed with the extended thrombolysis in cerebral infarction score (eTICI) and functional outcome with the modified Rankin Scale score (mRS) at 90 days. Sex-differences were assessed with multivariable regression analyses with adjustments for possible confounders. RESULTS 3180 patients were included (median age 72 years, 48% women). Clots in women were less often located in the intracranial internal carotid artery (ICA) (25%vs 28%, odds ratio (OR) 0.85;95% confidence interval: 0.73-1.00). CBS was similar between sexes (median 6, IQR 4-8). Intracranial (aOR 0.73;95% CI:0.62-0.87) and extracranial (aOR 0.64;95% CI:0.43-0.95) atherosclerosis was less prevalent in women. Vessel tortuosity was more frequent in women in the cervical ICA (aOR 1.89;95% CI:1.39-2.57) and women more often had severe elongation of the aortic arch (aOR 1.38;95% CI:1.00-1.91). ICA radius was smaller in women (2.3vs 2.5 mm, mean difference 0.22;95% CI:0.09-0.35) while M1 radius was essentially equal (1.6vs 1.7 mm, mean difference 0.09;95% CI:-0.02-0.21). Women had better collateral status (⩾50% filling in 62%vs 53% in men, aOR 1.48;95% CI:1.29-1.70). Finally, ASPECT scores were equal between women and men (median 9 in both sexes, IQR 8-10vs 9-10). Reperfusion rates were similar between women and men (acOR 0.94;95% CI:0.83-1.07). However, women less often reached functional independence than men (34%vs 46%, aOR 0.68;95% CI:0.53-0.86). DISCUSSION AND CONCLUSION On baseline imaging of this Dutch Registry, men and women with LVO mainly differ in vessel characteristics such as atherosclerotic burden, extracranial vessel tortuosity, and collateral status. These sex differences do not result in different reperfusion rates and are, therefore, not likely to explain the worse functional outcome in women after EVT.
Collapse
Affiliation(s)
- Anne van der Meij
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ghislaine Holswilder
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Louise E Bernsen
- Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, The Netherlands
| | - Hendrikus JA van Os
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health & Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeannette Hofmeijer
- Department of Neurology, Rijnstate Hospital, Arnhem, The Netherlands
- Department of Clinical Neurophysiology, University of Twente, Enschede, The Netherlands
| | | | - Jasper M Martens
- Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, The Netherlands
| | - Ido R van den Wijngaard
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, Haaglanden Medical Center, Den Haag, The Netherlands
| | - Hester F Lingsma
- Department of Public Health, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Praneeta R Konduri
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Charles BLM Majoie
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Diederik WJ Dippel
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nyika D Kruyt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul J Nederkoorn
- Department of Neurology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Marieke JH Wermer
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Hamam O, Gudenkauf J, Moustafa R, Cho A, Montes D, Sharara M, Moustafa A, Radmard M, Nabi M, Chen K, Sepehri S, Shin C, Mazumdar I, Kim M, Mohseni A, Malhotra A, Romero J, Yedavalli V. Hypoperfusion Intensity Ratio as an Indirect Imaging Surrogate in Patients With Anterior Circulation Large-Vessel Occlusion and Association of Baseline Characteristics With Poor Collateral Status. J Am Heart Assoc 2024; 13:e030897. [PMID: 39158547 DOI: 10.1161/jaha.123.030897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/15/2023] [Indexed: 08/20/2024]
Abstract
BACKGROUND Collateral status (CS) plays a crucial role in infarct growth rate, risk of postthrombectomy hemorrhage, and overall clinical outcomes in patients with acute ischemic stroke (AIS) secondary to anterior circulation large-vessel occlusions (LVOs). Hypoperfusion intensity ratio has been previously validated as an indirect noninvasive pretreatment imaging biomarker of CS. In addition to imaging, derangements in admission laboratory findings can also influence outcomes in patients with AIS-LVO. Therefore, our study aims to assess the relationship between admission laboratory findings, baseline characteristics, and CS, as assessed by hypoperfusion intensity ratio in patients with AIS-LVO. METHODS AND RESULTS In this retrospective study, consecutive patients presenting with AIS secondary to anterior circulation LVO who underwent pretreatment computed tomography perfusion were included. The computed tomography perfusion data processed by RAPID (Ischema View, Menlo Park, CA) generated the hypoperfusion intensity ratio. Binary logistic regression models were used to assess the relationship between patients' baseline characteristics, admission laboratory findings, and poor CS. A total of 221 consecutive patients with AIS-LVO between January 2017 and September 2022 were included in our study (mean±SD age, 67.0±15.8 years; 119 men [53.8%]). Multivariable logistic regression showed that patients with AIS caused by cardioembolic and cryptogenic causes (adjusted odds ratio [OR], 2.67; 95% CI, 1.20-5.97; P=0.016), those who presented with admission National Institutes of Health Stroke Scale score ≥12 (adjusted OR, 3.12; 95% CI, 1.61-6.04; P=0.001), and male patients (adjusted OR, 2.06; 95% CI, 1.13-3.77; P=0.018) were associated with poor CS. CONCLUSIONS Stroke caused by cardioembolic or cryptogenic causes, admission National Institutes of Health Stroke Scale score of ≥12, and male sex were associated with poor CS, as defined by hypoperfusion intensity ratio in the patients with AIS-LVO.
Collapse
Affiliation(s)
- Omar Hamam
- Department of Radiology, Massachusetts General Hospital Harvard Medical School Boston MA
| | - Julie Gudenkauf
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Rawan Moustafa
- Department of Cardiovascular Medicine Robert Wood Johnson Medical School New Brunswick NJ
- School of Arts and Sciences Rutgers University-Newark Newark NJ
| | - Andrew Cho
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Daniel Montes
- Radiology Resident University of Colorado, Anschutz Medical Campus Aurora CO
| | | | - Abdallah Moustafa
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Mahla Radmard
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Mehreen Nabi
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Kevin Chen
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Sadra Sepehri
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | | | - Ishan Mazumdar
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Minsoo Kim
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | - Alireza Mohseni
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| | | | - Javier Romero
- Department of Radiology, Massachusetts General Hospital Harvard Medical School Boston MA
| | - Vivek Yedavalli
- Department of Radiology and Radiological Science Johns Hopkins University School of Medicine Baltimore MD
| |
Collapse
|
6
|
Salim HA, Hamam O, Parilday G, Moustafa RA, Ghandour S, Rutgers M, Sharara M, Cho A, Mazumdar I, Radmard M, Shin C, Montes D, Malhotra A, Romero JM, Yedavalli V. Relative Cerebral Blood Flow as an Indirect Imaging Surrogate in Patients With Anterior Circulation Large Vessel Occlusion and Association of Baseline Characteristics With Poor Collateral Status. J Am Heart Assoc 2024; 13:e034581. [PMID: 39158542 DOI: 10.1161/jaha.124.034581] [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: 01/21/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND In acute ischemic stroke (AIS), collateral status (CS) is an important predictor of favorable outcomes in patients with AIS. Among quantitative cerebral perfusion parameters, relative cerebral blood flow (rCBF) is considered an accurate perfusion-based indicator of CS. This study investigated the relationship between admission laboratory values, baseline characteristics, and CS as assessed by rCBF in patients with AIS-large vessel occlusion. METHODS AND RESULTS In this retrospective multicenter study, consecutive patients presenting with AIS secondary to anterior circulation large vessel occlusion who underwent pretreatment computed tomography perfusion were included. The computed tomography perfusion data processed by RAPID (IschemaView, Menlo Park, CA) generated the rCBF. Binary logistic regression models assessed the relationship between patients' baseline characteristics, admission laboratory values, and poor CS. The primary outcome measure was the presence of poor CS, which was defined as rCBF <38% at a lesion size ≥27 mL. Between January 2017 and September 2022, there were 221 consecutive patients with AIS-large vessel occlusion included in our study (mean age 67.0±15.8 years, 119 men [53.8%]). Logistic regression showed that male sex (odds ratio [OR], 2.98 [1.59-5.59]; P=0.001), chronic kidney disease (OR, 5.18 [2.44-11.0]; P<0.001), admission National Institutes of Health Stroke Scale score ≥12 (OR, 5.17 [2.36-11.36]; P<0.001), and systolic blood pressure <140 (OR, 2.00 [1.07-3.76]; P=0.030) were associated with poor CS. CONCLUSIONS Higher stroke severity on admission with National Institutes of Health Stroke Scale score ≥12, systolic blood pressure <140, chronic kidney disease, and male sex are statistically significantly associated with poor CS in patients with AIS due to anterior circulation large vessel occlusion as defined by rCBF <38%.
Collapse
Affiliation(s)
| | | | | | | | | | - Moustafa Rutgers
- Rutgers University School of Arts and Sciences New Brunswick NJ USA
| | | | - Andrew Cho
- Johns Hopkins University School of Medicine Baltimore MD USA
| | - Ishan Mazumdar
- Johns Hopkins University School of Medicine Baltimore MD USA
| | | | | | - Daniel Montes
- University of Colorado Anschutz Medical Campus Aurora CA USA
| | | | | | | |
Collapse
|
7
|
Neubauer-Geryk J, Myśliwiec M, Bieniaszewski L. Gender-Related Difference in Skin Oxygenation in Young Patients with Uncomplicated Type 1 Diabetes. Biomedicines 2024; 12:1413. [PMID: 39061987 PMCID: PMC11274177 DOI: 10.3390/biomedicines12071413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Gender, through genetic, epigenetic and hormonal regulation, is an important modifier of the physiological mechanisms and clinical course of diseases. In diabetes mellitus, there are gender differences in incidence, prevalence, morbidity, and mortality. This disease also has an impact on the microvascular function. Therefore, this cross-sectional study was designed to investigate how gender affects the cutaneous microcirculation. We hypothesized that gender should be an important factor in the interpretation of capillaroscopy and transcutaneous oxygen saturation results. The study group consisted of 42 boys and 55 girls, uncomplicated diabetic pediatric patients. Females (F) and males (M) did not differ in terms of age, age at onset of diabetes, or diabetes duration. Furthermore, they did not differ in metabolic parameters. The comparison showed that group F had lower BP, higher pulse, and higher HR than group M. Group F had significantly lower creatinine and hemoglobin levels than group M. In children and adolescents with type 1 diabetes without complications, there was a gender difference in microcirculatory parameters. The resting transcutaneous partial pressure of oxygen was significantly higher in females than in males. However, there were no gender-related differences in basal capillaroscopic parameters or vascular reactivity during the PORH test. Our results indicate that studies investigating the structure and function of the microcirculation should consider the role of gender in addition to known cofactors such as puberty, body mass index, physical activity, and cigarette smoking.
Collapse
Affiliation(s)
- Jolanta Neubauer-Geryk
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Małgorzata Myśliwiec
- Department of Pediatrics, Diabetology and Endocrinology, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
| | - Leszek Bieniaszewski
- Clinical Physiology Unit, Medical Simulation Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| |
Collapse
|
8
|
Karapapak M, Ermis S, Aksöz Bolat P, Cingöz M, Erdim Ç, Özal E, Özal SA. Changes in retinal vascular density measured by optical coherence tomography angiography in patients with carotid artery stenosis after carotid artery stenting and angioplasty. Int Ophthalmol 2024; 44:128. [PMID: 38467951 DOI: 10.1007/s10792-024-03069-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
PURPOSE The aim of this study was to compare the effect of carotid artery stenting and angioplasty (CASA) on retinal vascular density (VD) in patients with severe carotid stenosis with a healthy control group and to evaluate using optical coherence tomography angiography (OCTA). METHODS For this prospective study, eyes on the operated side constituted the ipsilateral eye group, and the other eye constituted the contralateral eye group. 40 eyes of 40 patients with ipsilateral eye of carotisid artery stenosis (CAS), 34 eyes on contralateral side, and 30 healthy eyes (control group) were included in this study. We performed quantitative OCTA analyses of retinal VD changes, before and after CASA. The main outcome measures were the quantitative changes of VD of superficial capillary plexus (SCP) and deep capillary plexus (DCP). RESULTS We evaluated the VD of ipsilateral eyes and contralateral eyes separately before and after the procedure. All patients did not have visual symptoms. There was no significant difference in the VD of SCP in all groups before the procedure. No significant change was observed in all groups when the VD of the SCP was compared before and after the procedure. The VD of the DCP in the ipsilateral and contralateral group improved significantly after CASA. CONCLUSION OCTA could noninvasively detect retinal VD improvements after CASA in CAS patients. Quantitative changes in VD evaluated using OCTA are thought to be early indicators in the diagnosis of CAS and in the follow-up of treatment success.
Collapse
Affiliation(s)
- Murat Karapapak
- University of Health Sciences, Department of Ophthalmology, Basaksehir Cam and Sakura City Hospital, Basaksehir Olympic Boulevard Road Basaksehir, Istanbul, Turkey.
| | - Serhat Ermis
- University of Health Sciences, Department of Ophthalmology, Basaksehir Cam and Sakura City Hospital, Basaksehir Olympic Boulevard Road Basaksehir, Istanbul, Turkey
| | - Petek Aksöz Bolat
- University of Health Sciences, Department of Ophthalmology, Basaksehir Cam and Sakura City Hospital, Basaksehir Olympic Boulevard Road Basaksehir, Istanbul, Turkey
| | - Mehmet Cingöz
- University of Health Sciences, Department of Interventional Radiology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Çağrı Erdim
- University of Health Sciences, Department of Interventional Radiology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Ece Özal
- University of Health Sciences, Department of Ophthalmology, Basaksehir Cam and Sakura City Hospital, Basaksehir Olympic Boulevard Road Basaksehir, Istanbul, Turkey
| | - Sadık Altan Özal
- University of Health Sciences, Department of Ophthalmology, Basaksehir Cam and Sakura City Hospital, Basaksehir Olympic Boulevard Road Basaksehir, Istanbul, Turkey
| |
Collapse
|
9
|
Faber JE, Zhang H, Xenakis JG, Bell TA, Hock P, Pardo-Manuel de Villena F, Ferris MT, Rzechorzek W. Large differences in collateral blood vessel abundance among individuals arise from multiple genetic variants. J Cereb Blood Flow Metab 2023; 43:1983-2004. [PMID: 37572089 PMCID: PMC10676139 DOI: 10.1177/0271678x231194956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
Collateral blood flow varies greatly among humans for reasons that remain unclear, resulting in significant differences in ischemic tissue damage. A similarly large variation has also been found in mice that is caused by genetic background-dependent differences in the extent of collateral formation, termed collaterogenesis-a unique angiogenic process that occurs during development and determines collateral number and diameter in the adult. Previous studies have identified several quantitative trait loci (QTL) linked to this variation. However, understanding has been hampered by the use of closely related inbred strains that do not model the wide genetic variation present in the "outbred" human population. The Collaborative Cross (CC) multiparent mouse genetic reference panel was developed to address this limitation. Herein we measured the number and average diameter of cerebral collaterals in 60 CC strains, their 8 founder strains, 8 F1 crosses of CC strains selected for abundant versus sparse collaterals, and 2 intercross populations created from the latter. Collateral number evidenced 47-fold variation among the 60 CC strains, with 14% having poor, 25% poor-to-intermediate, 47% intermediate-to-good, and 13% good collateral abundance, that was associated with large differences in post-stroke infarct volume. Collateral number in skeletal muscle and intestine of selected high- and low-collateral strains evidenced the same relative abundance as in brain. Genome-wide mapping demonstrated that collateral abundance is a highly polymorphic trait. Subsequent analysis identified: 6 novel QTL circumscribing 28 high-priority candidate genes harboring putative loss-of-function polymorphisms (SNPs) associated with low collateral number; 335 predicted-deleterious SNPs present in their human orthologs; and 32 genes associated with vascular development but lacking protein coding variants. Six additional suggestive QTL (LOD > 4.5) were also identified in CC-wide QTL mapping. This study provides a comprehensive set of candidate genes for future investigations aimed at identifying signaling proteins within the collaterogenesis pathway whose variants potentially underlie genetic-dependent collateral insufficiency in brain and other tissues.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Neuroscience, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - James G Xenakis
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy A Bell
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Pablo Hock
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Wojciech Rzechorzek
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
10
|
Kaw A, Wu T, Starosolski Z, Zhou Z, Pedroza AJ, Majumder S, Duan X, Kaw K, Pinelo JEE, Fischbein MP, Lorenzi PL, Tan L, Martinez SA, Mahmud I, Devkota L, Taegtmeyer H, Ghaghada KB, Marrelli SP, Kwartler CS, Milewicz DM. Augmenting Mitochondrial Respiration in Immature Smooth Muscle Cells with an ACTA2 Pathogenic Variant Mitigates Moyamoya-like Cerebrovascular Disease. RESEARCH SQUARE 2023:rs.3.rs-3304679. [PMID: 37886459 PMCID: PMC10602100 DOI: 10.21203/rs.3.rs-3304679/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
ACTA2 pathogenic variants altering arginine 179 cause childhood-onset strokes due to moyamoya disease (MMD)-like occlusion of the distal internal carotid arteries. A smooth muscle cell (SMC)-specific knock-in mouse model (Acta2SMC-R179C/+) inserted the mutation into 67% of aortic SMCs, whereas explanted SMCs were uniformly heterozygous. Acta2R179C/+ SMCs fail to fully differentiate and maintain stem cell-like features, including high glycolytic flux, and increasing oxidative respiration (OXPHOS) with nicotinamide riboside (NR) drives the mutant SMCs to differentiate and decreases migration. Acta2SMC-R179C/+ mice have intraluminal MMD-like occlusive lesions and strokes after carotid artery injury, whereas the similarly treated WT mice have no strokes and patent lumens. Treatment with NR prior to the carotid artery injury attenuates the strokes, MMD-like lumen occlusions, and aberrant vascular remodeling in the Acta2SMC-R179C/+ mice. These data highlight the role of immature SMCs in MMD-associated occlusive disease and demonstrate that altering SMC metabolism to drive quiescence of Acta2R179C/+ SMCs attenuates strokes and aberrant vascular remodeling in the Acta2SMC-R179C/+ mice.
Collapse
Affiliation(s)
- Anita Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ting Wu
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Zbigniew Starosolski
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suravi Majumder
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Xueyan Duan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Kaveeta Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Jose E. E. Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sara A. Martinez
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Iqbal Mahmud
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laxman Devkota
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Heinrich Taegtmeyer
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ketan B. Ghaghada
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Sean P. Marrelli
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| |
Collapse
|
11
|
Rebchuk AD, Hill MD, Goyal M, Demchuk A, Coutts SB, Asdaghi N, Dowlatshahi D, Holodinsky JK, Fainardi E, Shankar J, Najm M, Rubiera M, Khaw AV, Qiu W, Menon BK, Field TS. Exploring sex differences for acute ischemic stroke clinical, imaging and thrombus characteristics in the INTERRSeCT study. J Cereb Blood Flow Metab 2023; 43:1803-1809. [PMID: 37459107 PMCID: PMC10581233 DOI: 10.1177/0271678x231189908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 10/17/2023]
Abstract
Women, especially following menopause, are known to have worse outcomes following acute ischemic stroke. One primary postulated biological mechanism for worse outcomes in older women is a reduction in the vasculoprotective effects of estrogen. Using the INTERRseCT cohort, a multicentre international observational cohort studying recanalization in acute ischemic stroke, we explored the effects of sex, and modifying effects of age, on neuroradiological predictors of recanalization including robustness of leptomeningeal collaterals, thrombus burden and thrombus permeability. Ordinal regression analyses were used to examine the relationship between sex and each of the neuroradiological markers. Further, we explored both multiplicative and additive interactions between age and sex. All patients (n = 575) from INTERRseCT were included. Mean age was 70.2 years (SD: 13.1) and 48.5% were women. In the unadjusted model, female sex was associated with better collaterals (OR 1.37, 95% CIs: 1.01-1.85), however this relationship was not significant after adjusting for age and relevant comorbidities. There were no significant interactions between age and sex. In a large prospective international cohort, we found no association between sex and radiological predictors of recanalization including leptomeningeal collaterals, thrombus permeability and thrombus burden.
Collapse
Affiliation(s)
- Alexander D Rebchuk
- Division of Neurosurgery, University of British Columbia, Vancouver, BC, Canada
| | - Michael D Hill
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Stroke Program, University of Calgary, Calgary, AB, Canada
| | - Mayank Goyal
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Stroke Program, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Andrew Demchuk
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Stroke Program, University of Calgary, Calgary, AB, Canada
| | - Shelagh B Coutts
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Stroke Program, University of Calgary, Calgary, AB, Canada
| | - Negar Asdaghi
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dar Dowlatshahi
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine (Neurology), University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jessalyn K Holodinsky
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Enrico Fainardi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Jai Shankar
- Department of Radiology, University of Manitoba, Winnipeg, MB, Canada
| | - Mohamed Najm
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marta Rubiera
- Neurology Department, Hospital Vall d’Hebron, Barcelona, Spain
| | - Alexander V Khaw
- Department of Clinical Neurosciences, University of Western Ontario, London, ON, Canada
| | - Wu Qiu
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Bijoy K Menon
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Calgary Stroke Program, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Thalia S Field
- Division of Neurology, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Vancouver Stroke Program, Vancouver, BC, Canada
| |
Collapse
|
12
|
Nagy D, Hricisák L, Walford GP, Lékai Á, Karácsony G, Várbíró S, Ungvári Z, Benyó Z, Pál É. Disruption of Vitamin D Signaling Impairs Adaptation of Cerebrocortical Microcirculation to Carotid Artery Occlusion in Hyperandrogenic Female Mice. Nutrients 2023; 15:3869. [PMID: 37764653 PMCID: PMC10534509 DOI: 10.3390/nu15183869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Vitamin D deficiency contributes to the pathogenesis of age-related cerebrovascular diseases, including ischemic stroke. Sex hormonal status may also influence the prevalence of these disorders, indicated by a heightened vulnerability among postmenopausal and hyperandrogenic women. To investigate the potential interaction between sex steroids and disrupted vitamin D signaling in the cerebral microcirculation, we examined the cerebrovascular adaptation to unilateral carotid artery occlusion (CAO) in intact, ovariectomized, and hyperandrogenic female mice with normal or functionally inactive vitamin D receptor (VDR). We also analyzed the morphology of leptomeningeal anastomoses, which play a significant role in the compensation. Ablation of VDR by itself did not impact the cerebrocortical adaptation to CAO despite the reduced number of pial collaterals. While ovariectomy did not undermine compensatory mechanisms following CAO, androgen excess combined with VDR inactivity resulted in prolonged hypoperfusion in the cerebral cortex ipsilateral to the occlusion. These findings suggest that the cerebrovascular consequences of disrupted VDR signaling are less pronounced in females, providing a level of protection even after ovariectomy. Conversely, even short-term androgen excess with lacking VDR signaling may lead to unfavorable outcomes of ischemic stroke, highlighting the complex interplay between sex steroids and vitamin D in terms of cerebrovascular diseases.
Collapse
Affiliation(s)
- Dorina Nagy
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - László Hricisák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Guillaume Peter Walford
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Ágnes Lékai
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Gábor Karácsony
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
| | - Szabolcs Várbíró
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary;
- Department of Obstetrics and Gynecology, University of Szeged, 6725 Szeged, Hungary
- Workgroup for Science Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
| | - Zoltán Ungvári
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, 1089 Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Éva Pál
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (L.H.); (G.P.W.); (Á.L.); (G.K.); (Z.B.)
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
13
|
Krausova V, Neumann D, Kraus J, Dostalova V, Dostal P. Sublingual microcirculation in healthy pediatric population using the sidestream dark-field imaging method. Clin Hemorheol Microcirc 2023; 85:163-171. [PMID: 37599527 DOI: 10.3233/ch-231851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
BACKGROUND The sidestream dark-field imaging method is used to study microcirculation. Normal values of sublingual microcirculation parameters in healthy children of different age and gender categories are unknown. OBJECTIVE The study's main goal was to determine normal values of selected parameters of sublingual microcirculation in healthy children of different age and gender categories. METHODS 40 healthy children were measured, ten aged 3-5.9 years, ten aged 6-10.9 years, ten aged 11-14.9 years, and ten aged 15-18.9 years. After recording the basic anthropometric parameters and vital functions, each volunteer had their microcirculation measured using an SDF probe placed sublingually. Three video clips were recorded and processed offline, and the three best and most stable parts of each were analyzed. RESULTS Total vascular density, small vessel density, proportion of perfused small vessels, perfused vessel density, perfused small vessel density, and DeBacker's score were significantly higher in females than in males. There were no differences between age groups in microcirculation parameters except MFI. CONCLUSIONS Age does not influence normal values of microcirculatory parameters. Female gender was associated with higher vessel density, perfused vessel density, and DeBacker's score. A suggestion of the normal range of microcirculatory parameters in healthy children is provided.
Collapse
Affiliation(s)
- Vlasta Krausova
- Department of Pediatrics, Masaryk Hospital, Krajska Zdravotni, Usti nad Labem, Czech Republic
- Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralové, Czech Republic
| | - David Neumann
- Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralové, Czech Republic
- Department of Pediatrics, Trutnov Hospital, Trutnov, Czech Republic
- Department of Pediatrics, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jaroslav Kraus
- Department od Orthopedics, Masaryk Hospital, Krajska Zdravotni, Usti nad Labem, Czech Republic
| | - Vlasta Dostalova
- Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Pavel Dostal
- Department of Anesthesiology and Intensive Care Medicine, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
14
|
Sabharwal R, Chapleau MW, Gerhold TD, Baumbach GL, Faraci FM. Plasticity of cerebral microvascular structure and mechanics during hypertension and following recovery of arterial pressure. Am J Physiol Heart Circ Physiol 2022; 323:H1108-H1117. [PMID: 36269650 PMCID: PMC9678426 DOI: 10.1152/ajpheart.00292.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022]
Abstract
Changes in vascular structure contribute to vascular events and loss of brain health. We examined changes in cerebral arterioles at the onset of hypertension and the hypothesis that alterations during hypertension would recover with the return of mean arterial pressure (MAP) to normal. MAP was measured with radiotelemetry in awake male C57BL/6J mice at baseline and during infusion of vehicle or angiotensin II (ANG II, 1.4 mg/kg/day using osmotic pumps) for 28 days, followed by a 28-day recovery. With ANG II treatment, MAP increased through day 28. On day 30, MAP began to recover, reaching levels not different from vehicle on day 37. We measured intravascular pressure, diameter, wall thickness (WT), wall:lumen ratio (W:L), cross-sectional area (CSA), and slope of the tangential elastic modulus (ET) in maximally dilated arterioles. Variables were similar in both groups at day 1, with no significant change with vehicle treatment. With ANG II treatment, CSA, WT, and W:L increased on days 7-28. Internal and external diameter was reduced at 14 and 28 days. ET versus wall stress was reduced on days 7-28. During recovery, the diameter remained at days 14 and 28 values, whereas other variables returned partly or completely to normal. Thus, CSA, WT, W:L, and ET versus wall stress changed rapidly during hypertension and recovered with MAP. In contrast, inward remodeling developed slowly and did not recover. This lack of recovery has mechanistic implications for the long-term impact of hypertension on vascular determinants of brain health.NEW & NOTEWORTHY Changes in vascular structure contribute to vascular events and loss of brain health. We examined the inherent structural plasticity of cerebral arterioles during and after a period of hypertension. Arteriolar wall thickness, diameter, wall-to-lumen ratio, and biological stiffness changed rapidly during hypertension and recovered with blood pressure. In contrast, inward remodeling developed slowly and did not recover. This lack of recovery of arteriolar diameter has implications for the long-term impact of hypertension on vascular determinants of brain health.
Collapse
Affiliation(s)
- Rasna Sabharwal
- Department of Internal Medicine, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
| | - Mark W Chapleau
- Department of Internal Medicine, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
| | - Thomas D Gerhold
- Department of Internal Medicine, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
| | - Gary L Baumbach
- Department of Pathology, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
| | - Frank M Faraci
- Department of Internal Medicine, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Francois M. Abboud Cardiovascular Center, University of Iowa, Iowa City, Iowa
| |
Collapse
|
15
|
Han MJ, Kim SJ. Clinical significance of asymmetric venous vasculature on minimum-intensity projection in patients with moyamoya disease. Medicine (Baltimore) 2022; 101:e31067. [PMID: 36254048 PMCID: PMC9575748 DOI: 10.1097/md.0000000000031067] [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: 11/26/2022] Open
Abstract
This study analyzed the clinical significance and characteristics of asymmetric venous blood flow in patients with Moyamoya disease (MMD) using minimum intensity projection (minIP) susceptibility-weighted imaging. The minIP views of 30 patients diagnosed with MMD were retrospectively analyzed using clinical features, brain magnetic resonance angiography, electroencephalography, and brain single-photon emission computed tomography (SPECT). Simultaneously, differences between patients with acute cerebral infarction and non-MMD causes were analyzed. Twelve (40.0%) of the 30 patients had asymmetrical venous flow, which is usually seen in patients with acute cerebral infarction (P = .146). They also had significantly higher Suzuki stages than symmetric patients (P = .014), with five (41.7%) and three (25.0%) of them in stages 4 and 5, respectively. When the Suzuki stages of both hemispheres were different, more veins were found in the stenotic hemisphere (88.9%). Brain SPECT showed more severe hypoperfusion on the side with prominent vascularity in the minIP view (100.0%). Additionally, asymmetric blood flow was observed in 66.7% of the patients with cerebral infarction caused by MMD, whereas only 11.1% of the children with cerebral infarction caused by non-MMD had asymmetry (P = .005). Patients with MMD showed asymmetric hypointensity of the cortical veins with a minIP appearance. The venous structure showed greater signal loss on SWI and was more prominent in the hemisphere where stenosis was advanced or infarction occurred in other examinations. Cerebral infarction in patients with MMD tended to occur with asymmetrically prominent venous patterns with damaged areas in minIP images, which had distinct characteristics from those of patients without MMD.
Collapse
Affiliation(s)
- Min Jeong Han
- Department of Pediatrics, Jeonbuk National University Medical School, Jeonbuk, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonbuk National University Medical School, Jeonbuk, Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonbuk National University Medical School, Jeonbuk, Korea
| | - Sun Jun Kim
- Department of Pediatrics, Jeonbuk National University Medical School, Jeonbuk, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonbuk National University Medical School, Jeonbuk, Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonbuk National University Medical School, Jeonbuk, Korea
- *Correspondence: Sun Jun Kim, Department of Pediatrics, Jeonbuk National University Medical School, 20 Geonjiro, Deokjingu, Jeonju, Jeonbuk, 54907, South Korea (e-mail: )
| |
Collapse
|
16
|
Uniken Venema SM, Dankbaar JW, van der Lugt A, Dippel DWJ, van der Worp HB. Cerebral Collateral Circulation in the Era of Reperfusion Therapies for Acute Ischemic Stroke. Stroke 2022; 53:3222-3234. [PMID: 35938420 DOI: 10.1161/strokeaha.121.037869] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clinical outcomes of patients with acute ischemic stroke depend in part on the extent of their collateral circulation. A good collateral circulation has also been associated with greater benefit of intravenous thrombolysis and endovascular treatment. Treatment decisions for these reperfusion therapies are increasingly guided by a combination of clinical and imaging parameters, particularly in later time windows. Computed tomography and magnetic resonance imaging enable a rapid assessment of both the collateral extent and cerebral perfusion. Yet, the role of the collateral circulation in clinical decision-making is currently limited and may be underappreciated due to the use of rather coarse and rater-dependent grading methods. In this review, we discuss determinants of the collateral circulation in patients with acute ischemic stroke, report on commonly used and emerging neuroimaging techniques for assessing the collateral circulation, and discuss the therapeutic and prognostic implications of the collateral circulation in relation to reperfusion therapies for acute ischemic stroke.
Collapse
Affiliation(s)
- Simone M Uniken Venema
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, the Netherlands. (S.M.U.V., H.B.v.d.W.)
| | - Jan Willem Dankbaar
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, the Netherlands. (J.W.D.)
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center Rotterdam, the Netherlands. (A.v.d.L.)
| | - Diederik W J Dippel
- Department of Neurology, Erasmus Medical Center Rotterdam, the Netherlands. (D.W.J.D.)
| | - H Bart van der Worp
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, the Netherlands. (S.M.U.V., H.B.v.d.W.)
| |
Collapse
|
17
|
Xu Q, Sun H, Yi Q. Association Between Retinal Microvascular Metrics Using Optical Coherence Tomography Angiography and Carotid Artery Stenosis in a Chinese Cohort. Front Physiol 2022; 13:824646. [PMID: 35721537 PMCID: PMC9204184 DOI: 10.3389/fphys.2022.824646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: The main aim was to investigate the association between retinal microvascular metrics using optical coherence tomography angiography (OCTA) and carotid artery stenosis (CAS) in an aging Chinese cohort.Methods: In this cross-sectional and observational study, 138 eyes of 138 participants were examined. Indices of the microcirculation measured by OCTA included mean vessel density (VD), skeleton density (SD), vessel diameter index (VDI), fractal dimension (FD) and foveal avascular zone (FAZ) of the superficial retinal layer (SRL) and deep retinal layer (DRL), and peripapillary vessel caliber. The correlation of these indices with the carotid atherosclerotic lesions including carotid intima media thickness (CIMT) and common carotid artery (CCA) plaque was assessed.Results: A total of 72 of 138 eyes demonstrated an increased (≥1 mm) CIMT, and 32 of the eyes presented common carotid plaques. Macular VD, SD, and FD were decreased with the increasing CCA caliber diameter (p < 0.05, respectively). Superficial and deep macular FDs were negatively associated with CIMT as well as the existence of CCA plaques (p < 0.05, respectively).Conclusion: Changes in retinal microvasculature accessed by OCTA may be used as one of the non-invasive early indicators to monitor asymptomatic CAS.
Collapse
Affiliation(s)
- Qian Xu
- Qilu Hospital, Shandong University, Jinan, China
- Tai’an City Central Hospital, Tai’an, China
| | - Hongyi Sun
- Qilu Hospital, Shandong University, Jinan, China
| | - Qu Yi
- Qilu Hospital, Shandong University, Jinan, China
- *Correspondence: Qu Yi,
| |
Collapse
|
18
|
Lagebrant C, Ramgren B, Hassani Espili A, Marañon A, Kremer C. Sex Differences in Collateral Circulation and Outcome After Mechanical Thrombectomy in Acute Ischemic Stroke. Front Neurol 2022; 13:878759. [PMID: 35665053 PMCID: PMC9160377 DOI: 10.3389/fneur.2022.878759] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/19/2022] [Indexed: 11/23/2022] Open
Abstract
Background Collateral circulation is known to lead to smaller infarct volume and better functional outcome after mechanical thrombectomy (MT), but studies examining sex differences in collateral circulation are scarce. The aim of this study was to investigate if collateral circulation has a different impact on outcome in women and men. Methods A single-center retrospective study of 487 patients (230 men and 257 women) treated with MT for acute ischemic stroke in the anterior cerebral circulation. Collateral circulation was assessed on computed tomography angiography images. The outcome was evaluated at 90 days according to the modified Rankin Scale (mRS). Results Women were older, median age 76 years (IQR 68-83) vs. 71 years (IQR 63–78). Stroke severity and time to recanalization were comparable. More women had moderate or good collaterals in 58.4 vs. 47.0% for men (p = 0.01). Among patients with moderate and good collaterals significantly more men (61%) were functionally independent (mRS 0–2) than women (41.5%) (p = < 0.01). This difference remained significant after correcting for age by linear weighting, 60.4 vs. 46.8% (p = 0.03). Conclusion Women had better collateral flow but showed worse functional outcomes, while good collateral flow led to better outcomes in men, even after correcting for age. Further clinical studies on peri- and post-interventional care, factors affecting recovery after hospital discharge as well as basic research on the neurovascular unit are needed to find modifiable targets to improve clinical outcomes for women.
Collapse
Affiliation(s)
| | - Birgitta Ramgren
- Department of Diagnostic Radiology, Neuroradiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | | | - Christine Kremer
- Neurology Department, Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Lund, Sweden
- *Correspondence: Christine Kremer
| |
Collapse
|
19
|
Lee S, Jiang B, Wintermark M, Mlynash M, Christensen S, Sträter R, Broocks G, Grams A, Dorn F, Nikoubashman O, Kaiser D, Morotti A, Jensen-Kondering U, Trenkler J, Möhlenbruch M, Fiehler J, Wildgruber M, Kemmling A, Psychogios M, Sporns PB. Cerebrovascular Collateral Integrity in Pediatric Large Vessel Occlusion: Analysis of the Save ChildS Study. Neurology 2022; 98:e352-e363. [PMID: 34795051 DOI: 10.1212/wnl.0000000000013081] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/22/2021] [Accepted: 11/04/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Robust cerebrovascular collaterals in adult patients with large vessel occlusion stroke have been associated with longer treatment windows, better recanalization rates, and improved outcomes, but the role of collaterals in pediatric stroke is not known. The primary aim was to determine whether favorable collaterals correlated with better radiographic and clinical outcomes in children with ischemic stroke who underwent thrombectomy. METHODS This study analyzed a subset of children enrolled in SaveChildS, a retrospective, multicenter, observational cohort study of 73 pediatric patients with stroke who underwent thrombectomy between 2000 and 2018 at 27 US and European centers. Included patients had baseline angiographic imaging and follow-up modified Rankin Scale scores available for review. Posterior circulation occlusions were excluded. Cerebrovascular collaterals were graded on acute neuroimaging by 2 blinded neuroradiologists according to the Tan collateral score, in which favorable collaterals are defined as >50% filling and unfavorable collaterals as <50% filling distal to the occluded vessel. Collateral status was correlated with clinical and neuroimaging characteristics and outcomes. Between-group comparisons were performed with the Wilcoxon rank-sum test for continuous variables or Fisher exact test for binary variables. RESULTS Thirty-three children (mean age 10.9 [SD ±4.9]) years were included; 14 (42.4%) had favorable collaterals. Median final stroke volume as a percent of total brain volume (TBV) was significantly lower in patients with favorable collaterals (1.35% [interquartile range (IQR) 1.14%-3.76%] vs 7.86% [IQR 1.54%-11.07%], p = 0.049). Collateral status did not correlate with clinical outcome, infarct growth, or final Alberta Stroke Program Early CT Score (ASPECTS) in our cohort. Patients with favorable collaterals had higher baseline ASPECTS (7 [IQR 6-8] vs 5.5 [4-6], p = 0.006), smaller baseline ischemic volume (1.57% TBV [IQR 1.09%-2.29%] vs 3.42% TBV [IQR 1.26%-5.33%], p = 0.035), and slower early infarct growth rate (2.4 mL/h [IQR 1.5-5.1 mL/h] vs 10.4 mL/h [IQR 3.0-30.7 mL/h], p = 0.028). DISCUSSION Favorable collaterals were associated with smaller final stroke burden and slower early infarct growth rate but not with better clinical outcome in our study. Prospective studies are needed to determine the impact of collaterals in childhood stroke. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that in children with ischemic stroke undergoing thrombectomy, favorable collaterals were associated with improved radiographic outcomes but not with better clinical outcomes.
Collapse
Affiliation(s)
- Sarah Lee
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland.
| | - Bin Jiang
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Max Wintermark
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Michael Mlynash
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Soren Christensen
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Ronald Sträter
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Gabriel Broocks
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Astrid Grams
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Franziska Dorn
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Omid Nikoubashman
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Daniel Kaiser
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Andrea Morotti
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Ulf Jensen-Kondering
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Johannes Trenkler
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Markus Möhlenbruch
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Jens Fiehler
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Moritz Wildgruber
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - André Kemmling
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Marios Psychogios
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| | - Peter B Sporns
- From the Department of Neurology & Neurological Sciences, Stanford Stroke Center (S.L., M. Mlynash, S.C.), Department of Neurology & Neurological Sciences (S.L.), Division of Child Neurology, and Department of Radiology (B.J., M. Wintermark), Division of Neuroradiology, Stanford University School of Medicine, CA; Department of Pediatrics (R.S.), University Hospital of Muenster; Department of Diagnostic and Interventional Neuroradiology (G.B., J.F., P.B.S.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Neuroradiology (A.G.), Medical University of Innsbruck, Austria; Department of Neuroradiology (F.D.), University Hospital Bonn; Department of Neuroradiology (O.N.), RWTH Aachen University; Department of Neuroradiology (D.K.), University Hospital Carl Gustav Carus, Dresden, Germany; ASST Valcamonica (A.M.), UOSD Neurology, Esine (BS), Brescia, Italy; Department of Radiology and Neuroradiology (U.J.-K.), University Hospital of Schleswig-Holstein, Campus Kiel; Institute of Neuroradiology (U.J.-K.), UKSH Campus Lübeck, Germany; Department of Neuroradiology (J.T.), Kepler University Hospital, Johannes Kepler University Linz, Austria; Department of Neuroradiology (M. Möhlenbruch), Heidelberg University Hospital; Department of Radiology (M. Wildgruber), University Hospital, LMU Munich; Department of Neuroradiology (A.K.), Marburg University Hospital, Germany; and Department of Neuroradiology (M.P., P.B.S.), Clinic for Radiology & Nuclear Medicine, University Hospital Basel, Switzerland
| |
Collapse
|
20
|
Determinants of Leptomeningeal Collateral Status Variability in Ischemic Stroke Patients. Can J Neurol Sci 2021; 49:767-773. [PMID: 34585652 DOI: 10.1017/cjn.2021.226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Collateral status is an indicator of a favorable outcome in stroke. Leptomeningeal collaterals provide alternative routes for brain perfusion following an arterial occlusion or flow-limiting stenosis. Using a large cohort of ischemic stroke patients, we examined the relative contribution of various demographic, laboratory, and clinical variables in explaining variability in collateral status. METHODS Patients with acute ischemic stroke in the anterior circulation were enrolled in a multi-center hospital-based observational study. Intracranial occlusions and collateral status were identified and graded using multiphase computed tomography angiography. Based on the percentage of affected territory filled by collateral supply, collaterals were graded as either poor (0-49%), good (50-99%), or optimal (100%). Between-group differences in demographic, laboratory, and clinical factors were explored using ordinal regression models. Further, we explored the contribution of measured variables in explaining variance in collateral status. RESULTS 386 patients with collateral status classified as poor (n = 64), good (n = 125), and optimal (n = 197) were included. Median time from symptom onset to CT was 120 (IQR: 78-246) minutes. In final multivariable model, male sex (OR 1.9, 95% CIs [1.2, 2.9], p = 0.005) and leukocytosis (OR 1.1, 95% CIs [1.1, 1.2], p = 0.001) were associated with poor collaterals. Measured variables only explained 44.8-53.0% of the observed between-patient variance in collaterals. CONCLUSION Male sex and leukocytosis are associated with poorer collaterals. Nearly half of the variance in collateral flow remains unexplained and could be in part due to genetic differences.
Collapse
|
21
|
Derraz I, Abdelrady M, Gaillard N, Ahmed R, Cagnazzo F, Dargazanli C, Lefevre PH, Corti L, Riquelme C, Mourand I, Gascou G, Bonafe A, Arquizan C, Costalat V. White Matter Hyperintensity Burden and Collateral Circulation in Large Vessel Occlusion Stroke. Stroke 2021; 52:3848-3854. [PMID: 34517773 DOI: 10.1161/strokeaha.120.031736] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE White matter hyperintensity (WMH), a marker of chronic cerebral small vessel disease, might impact the recruitment of leptomeningeal collaterals. We aimed to assess whether the WMH burden is associated with collateral circulation in patients treated by endovascular thrombectomy for anterior circulation acute ischemic stroke. METHODS Consecutive acute ischemic stroke due to anterior circulation large vessel occlusion and treated with endovascular thrombectomy from January 2015 to December 2017 were included. WMH volumes (periventricular, deep, and total) were assessed by a semiautomated volumetric analysis on fluid-attenuated inversion recovery-magnetic resonance imaging. Collateral status was graded on baseline catheter angiography using the American Society of Interventional and Therapeutic Neuroradiology/Society of Interventional Radiology grading system (good when ≥3). We investigated associations of WMH burden with collateral status. RESULTS A total of 302 patients were included (mean age, 69.1±19.4 years; women, 55.6%). Poor collaterals were observed in 49.3% of patients. Median total WMH volume was 3.76 cm3 (interquartile range, 1.09-11.81 cm3). The regression analyses showed no apparent relationship between WMH burden and the collateral status measured at baseline angiography (adjusted odds ratio, 0.987 [95% CI, 0.971-1.003]; P=0.12). CONCLUSIONS WMH burden exhibits no overt association with collaterals in large vessel occlusive stroke.
Collapse
Affiliation(s)
- Imad Derraz
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Mohamed Abdelrady
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Nicolas Gaillard
- Department of Neurology (N.G., L.C., I.M., C.A.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Raed Ahmed
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Federico Cagnazzo
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Cyril Dargazanli
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Pierre-Henri Lefevre
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Lucas Corti
- Department of Neurology (N.G., L.C., I.M., C.A.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Carlos Riquelme
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Isabelle Mourand
- Department of Neurology (N.G., L.C., I.M., C.A.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Gregory Gascou
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Alain Bonafe
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Caroline Arquizan
- Department of Neurology (N.G., L.C., I.M., C.A.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| | - Vincent Costalat
- Department of Neuroradiology (I.D., M.A., R.A., F.C., C.D., P.-H.L., C.R., G.G., A.B., V.C.), Hôpital Gui de Chauliac, Montpellier University Medical Center, Montpellier, France
| |
Collapse
|
22
|
Lin J, Cheng Z, Shi Y, Cai X, Huang L. Evaluating the Velocity and Extent of Cortical Venous Filling in Patients With Severe Middle Cerebral Artery Stenosis or Occlusion. Front Neurol 2021; 12:610658. [PMID: 33897584 PMCID: PMC8060485 DOI: 10.3389/fneur.2021.610658] [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: 09/26/2020] [Accepted: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the velocity and extent of cortical venous filling (CVF) and its association with clinical manifestations in patients with severe stenosis or occlusion of the middle cerebral artery (MCA) using dynamic computed tomography angiography (CTA). Methods: Fifty-eight patients (36 symptomatic and 22 asymptomatic) with severe unilateral stenosis (≥70%) or occlusion of the MCA M1 segment who underwent dynamic CTA were included. Collateral status, antegrade flow, and CVF of each patient were observed using dynamic CTA. Three types of cortical veins were selected to observe the extent of CVF, and the absence of CVF (CVF-) was recorded. Based on the appearance of CVF in the superior sagittal sinus, instances of CVF, including early (CVF1), peak (CVF2), and late (CVF3) venous phases, were recorded. The differences in CVF times between the affected and contralateral hemispheres were represented as rCVFs, and CVF velocity was defined compared to the median time of each rCVF. Results: All CVF times in the affected hemisphere were longer than those in the contralateral hemisphere (p < 0.05). Patients with symptomatic MCA stenosis had more ipsilateral CVF- (p = 0.02) and more delayed CVF at rCVF2 and rCVF21 (rCVF2-rCVF1) (p = 0.03 and 0.001, respectively) compared to those with asymptomatic MCA stenosis. For symptomatic patients, fast CVF at rCVF21 was associated with poor collateral status (odds ratio [OR] 6.42, 95% confidence interval [CI] 1.37-30.05, p = 0.02), and ipsilateral CVF- in two cortical veins was associated with poor 3-month outcomes (adjusted OR 0.025, 95% CI 0.002-0.33, p = 0.005). Conclusions: Complete and fast CVF is essential for patients with symptomatic MCA stenosis or occlusion. The clinical value of additional CVF assessment should be explored in future studies to identify patients with severe MCA stenosis or occlusion at a higher risk of stroke occurrence and poor recovery.
Collapse
Affiliation(s)
- Jia'Xing Lin
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zhong'Yuan Cheng
- Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying'Ying Shi
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiang'Ran Cai
- Medical Imaging Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Li'An Huang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
23
|
Faber JE, Storz JF, Cheviron ZA, Zhang H. High-altitude rodents have abundant collaterals that protect against tissue injury after cerebral, coronary and peripheral artery occlusion. J Cereb Blood Flow Metab 2021; 41:731-744. [PMID: 32703056 PMCID: PMC7983333 DOI: 10.1177/0271678x20942609] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/03/2020] [Accepted: 06/22/2020] [Indexed: 12/22/2022]
Abstract
Collateral number/density varies widely in brain and other tissues among strains of Mus musculus mice due to differences in genetic background. Recent studies have shown that prolonged exposure to reduced atmospheric oxygen induces additional collaterals to form, suggesting that natural selection may favor increased collaterals in populations native to high-altitude. High-altitude guinea pigs (Cavia) and deer mice (Peromyscus) were compared with lowland species of Peromyscus, Mus and Rattus (9 species/strains examined). Collateral density, diameter and other morphometrics were measured in brain where, importantly, collateral abundance reflects that in other tissues of the same individual. Guinea pigs and high-altitude deer mice had a greater density of pial collaterals than lowlanders. Consistent with this, guinea pigs and highlander mice evidenced complete and 80% protection against stroke, respectively. They also sustained significantly less ischemia in heart and lower extremities after arterial occlusion. Vessels of the circle of Willis, including the communicating collateral arteries, also exhibited unique features in the highland species. Our findings support the hypothesis that species native to high-altitude have undergone genetic selection for abundant collaterals, suggesting that besides providing protection in obstructive disease, collaterals serve a physiological function to optimize oxygen delivery to meet oxygen demand when oxygen is limiting.
Collapse
Affiliation(s)
- James E Faber
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jay F Storz
- School of Biological Sciences, University of Nebraska, Lincoln, NE, USA
| | - Zachary A Cheviron
- Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
24
|
PHACTR1 genetic variability is not critical in small vessel ischemic disease patients and PcomA recruitment in C57BL/6J mice. Sci Rep 2021; 11:6072. [PMID: 33727568 PMCID: PMC7966789 DOI: 10.1038/s41598-021-84919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 02/04/2021] [Indexed: 11/29/2022] Open
Abstract
Recently, several genome-wide association studies identified PHACTR1 as key locus for five diverse vascular disorders: coronary artery disease, migraine, fibromuscular dysplasia, cervical artery dissection and hypertension. Although these represent significant risk factors or comorbidities for ischemic stroke, PHACTR1 role in brain small vessel ischemic disease and ischemic stroke most important survival mechanism, such as the recruitment of brain collateral arteries like posterior communicating arteries (PcomAs), remains unknown. Therefore, we applied exome and genome sequencing in a multi-ethnic cohort of 180 early-onset independent familial and apparently sporadic brain small vessel ischemic disease and CADASIL-like Caucasian patients from US, Portugal, Finland, Serbia and Turkey and in 2 C57BL/6J stroke mouse models (bilateral common carotid artery stenosis [BCCAS] and middle cerebral artery occlusion [MCAO]), characterized by different degrees of PcomAs patency. We report 3 very rare coding variants in the small vessel ischemic disease-CADASIL-like cohort (p.Glu198Gln, p.Arg204Gly, p.Val251Leu) and a stop-gain mutation (p.Gln273*) in one MCAO mouse. These coding variants do not cluster in PHACTR1 known pathogenic domains and are not likely to play a critical role in small vessel ischemic disease or brain collateral circulation. We also exclude the possibility that copy number variants (CNVs) or a variant enrichment in Phactr1 may be associated with PcomA recruitment in BCCAS mice or linked to diverse vascular traits (cerebral blood flow pre-surgery, PcomA size, leptomeningeal microcollateral length and junction density during brain hypoperfusion) in C57BL/6J mice, respectively. Genetic variability in PHACTR1 is not likely to be a common susceptibility factor influencing small vessel ischemic disease in patients and PcomA recruitment in C57BL/6J mice. Nonetheless, rare variants in PHACTR1 RPEL domains may influence the stroke outcome and are worth investigating in a larger cohort of small vessel ischemic disease patients, different ischemic stroke subtypes and with functional studies.
Collapse
|
25
|
Regenhardt RW, Etherton MR, Das AS, Schirmer MD, Hirsch JA, Stapleton CJ, Patel AB, Leslie-Mazwi TM, Rost NS. Infarct Growth despite Endovascular Thrombectomy Recanalization in Large Vessel Occlusive Stroke. J Neuroimaging 2021; 31:155-164. [PMID: 33119954 PMCID: PMC8365346 DOI: 10.1111/jon.12796] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/14/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Endovascular thrombectomy (EVT) has revolutionized large vessel occlusion stroke care. However, not all patients with good endovascular results achieve good outcomes. We sought to understand the clinical significance of magnetic resonance imaging defined infarct growth despite adequate reperfusion and identify associated clinical and radiographic variables. METHODS History, presentation, treatments, and outcomes for consecutive EVT patients at a referral center were collected. Adequate reperfusion was defined as thrombolysis in cerebral infarction (TICI) score 2b-3. Region-specific infarct volumes in white matter, cortex, and basal ganglia were determined on diffusion-weighted imaging. Infarct growth was defined as post-EVT minus pre-EVT volume. Good outcome was defined as 90-day modified Rankin Scale ≤2. RESULTS Forty-four patients with adequate reperfusion were identified with median age 72 years; 64% were women. Each region showed infarct growth: white matter (median pre-EVT 7 cubic centimeters [cc], post-EVT 16 cc), cortex (4 cc, 15 cc), basal ganglia (2 cc, 4 cc), total (20 cc, 39 cc). In multivariable regression, total infarct growth independently decreased the odds of good outcome (odds ratio = .946, 95% CI = .897, .998). Further multivariable analyses for determinants of infarct growth identified female sex was associated with less total growth (β = -.294, P = .042), TICI 3 was associated with less white matter growth (β = -.277, P = .048) and cortical growth (β = -.335, P = .017), and both female sex (β = -.332, P = .015) and coronary disease (β = -.337, P = .015) were associated with less cortical growth. CONCLUSIONS Infarct growth occurred despite adequate reperfusion, disproportionately in the cortex, and independently decreased the odds of good outcome. Infarct growth occurred while patients were hospitalized and may represent a therapeutic target. Potential determinants of region-specific infarct growth were identified that require confirmation in larger studies.
Collapse
Affiliation(s)
- Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School
| | - Mark R Etherton
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Alvin S Das
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Markus D Schirmer
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Joshua A Hirsch
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School
| | | | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School
| | - Thabele M Leslie-Mazwi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School
| | - Natalia S Rost
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| |
Collapse
|
26
|
Aghajanian A, Zhang H, Buckley BK, Wittchen ES, Ma WY, Faber JE. Decreased inspired oxygen stimulates de novo formation of coronary collaterals in adult heart. J Mol Cell Cardiol 2021; 150:1-11. [PMID: 33038388 PMCID: PMC7855913 DOI: 10.1016/j.yjmcc.2020.09.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 01/18/2023]
Abstract
RATIONALE Collateral vessels lessen myocardial ischemia when acute or chronic coronary obstruction occurs. It has long been assumed that although native (pre-existing) collaterals enlarge in obstructive disease, new collaterals do not form in the adult. However, the latter was recently shown to occur after coronary artery ligation. Understanding the signals that drive this process is challenged by the difficulty in studying collateral vessels directly and the complex milieu of signaling pathways, including cell death, induced by ligation. Herein we show that hypoxemia alone is capable of inducing collateral vessels to form and that the novel gene Rabep2 is required. OBJECTIVE Hypoxia stimulates angiogenesis during embryonic development and in pathological states. We hypothesized that hypoxia also stimulates collateral formation in adult heart by a process that involves RABEP2, a recently identified protein required for formation of collateral vessels during development. METHODS AND RESULTS Exposure of mice to reduced FiO2 induced collateral formation that resulted in smaller infarctions following LAD ligation and that reversed on return to normoxia. Deletion of Rabep2 or knockdown of Vegfa inhibited formation. Hypoxia upregulated Rabep2, Vegfa and Vegfr2 in heart and brain microvascular endothelial cells (HBMVECs). Knockdown of Rabep2 impaired migration of HBMVECs. In contrast to systemic hypoxia, deletion of Rabep2 did not affect collateral formation induced by ischemic injury caused by LAD ligation. CONCLUSIONS Hypoxia induced formation of coronary collaterals by a process that required VEGFA and RABEP2, proteins also required for collateral formation during development. Knockdown of Rabep2 impaired cell migration, providing one potential mechanism for RABEP2's role in collateral formation. This appears specific to hypoxia, since formation after acute ischemic injury was unaffected in Rabep2-/- mice. These findings provide a novel model for studying coronary collateral formation, and demonstrate that hypoxia alone can induce new collaterals to form in adult heart.
Collapse
Affiliation(s)
- Amir Aghajanian
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Hua Zhang
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Brian K Buckley
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Erika S Wittchen
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - Willa Y Ma
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America
| | - James E Faber
- Department of Cell Biology and Physiology and the McAllister Heart Institute, University of North Carolina at Chapel Hill, United States of America.
| |
Collapse
|
27
|
Watanabe N, Noda Y, Nemoto T, Iimura K, Shimizu T, Hotta H. Cerebral artery dilation during transient ischemia is impaired by amyloid β deposition around the cerebral artery in Alzheimer's disease model mice. J Physiol Sci 2020; 70:57. [PMID: 33302862 PMCID: PMC10718030 DOI: 10.1186/s12576-020-00785-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/21/2020] [Indexed: 11/10/2022]
Abstract
Transient ischemia is an exacerbation factor of Alzheimer's disease (AD). We aimed to examine the influence of amyloid β (Aβ) deposition around the cerebral (pial) artery in terms of diameter changes in the cerebral artery during transient ischemia in AD model mice (APPNL-G-F) under urethane anesthesia. Cerebral vasculature and Aβ deposition were examined using two-photon microscopy. Cerebral ischemia was induced by transient occlusion of the unilateral common carotid artery. The diameter of the pial artery was quantitatively measured. In wild-type mice, the diameter of arteries increased during occlusion and returned to their basal diameter after re-opening. In AD model mice, the artery response during occlusion differed depending on Aβ deposition sites. Arterial diameter changes at non-Aβ deposition site were similar to those in wild-type mice, whereas they were significantly smaller at Aβ deposition site. The results suggest that cerebral artery changes during ischemia are impaired by Aβ deposition.
Collapse
Affiliation(s)
- Nobuhiro Watanabe
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yoshihiro Noda
- Animal Facility, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Taeko Nemoto
- Animal Facility, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Kaori Iimura
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Takahiko Shimizu
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Aichi, 474-8511, Japan
| | - Harumi Hotta
- Department of Autonomic Neuroscience, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo, 173-0015, Japan.
| |
Collapse
|
28
|
Wen H, Lv M. Correlation analysis between serum procalcitonin and infarct volume in young patients with acute cerebral infarction. Neurol Sci 2020; 42:3189-3196. [PMID: 33108576 DOI: 10.1007/s10072-020-04856-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/22/2020] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To detect the serum procalcitonin (PCT) level and determine its relationship with the infarct volume in young patients with acute cerebral infarction. METHODS According to the infarct volume, young patients with acute cerebral infarction were divided into large group, intermediate group, and small group. The severity of clinical symptoms was determined according to the National Institute of Health Stroke Scale (NIHSS) score. Healthy young people were selected as the control group. Serum PCT levels were measured. The relationship among PCT, volume, and NIHSS score was analyzed. RESULTS PCT in observation group was significantly higher than that in control group (t = 6.879, P = 0.011), and PCT in severe group was significantly higher than in mild group (t = 6.978, P = 0.016). PCT in large cerebral infarction group was higher than that in intermediate and small-size infarct group (P = 0.0036 and P < 0.0001, respectively), and PCT in intermediate cerebral infarction group was higher than that in small-size infarct group (P = 0.0024). In observation group, the PCT level was positively correlated with both NIHSS (r = 0.793, P = 0.022) and infarction volume (r = 0.649, P = 0.007). CONCLUSION The level of PCT in young patients with acute cerebral infarction may be related to the inflammatory reaction of the cerebral artery and positively related to the volume of cerebral infarction and NIHSS score. To some extent, PCT concentration can predict the disease severity of acute cerebral infarction.
Collapse
Affiliation(s)
- Huijun Wen
- Department of Neurology, Baoji Municipal Central Hospital, 8 Jiangtan Road, Baoji, 721008, Shaanxi, People's Republic of China
| | - Maikou Lv
- Department of Neurology, Baoji Municipal Central Hospital, 8 Jiangtan Road, Baoji, 721008, Shaanxi, People's Republic of China.
| |
Collapse
|
29
|
Yanagisawa T, Zhang H, Suzuki T, Kamio Y, Takizawa T, Morais A, Chung DY, Qin T, Murayama Y, Faber JE, Patel AB, Ayata C. Sex and Genetic Background Effects on the Outcome of Experimental Intracranial Aneurysms. Stroke 2020; 51:3083-3094. [PMID: 32912097 DOI: 10.1161/strokeaha.120.029651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Intracranial aneurysm formation and rupture risk are, in part, determined by genetic factors and sex. To examine their role, we compared 3 mouse strains commonly used in cerebrovascular studies in a model of intracranial aneurysm formation and rupture. METHODS Intracranial aneurysms were induced in male CD1 (Crl:CD1[ICR]), male and female C57 (C57BL/6NCrl), and male 129Sv (129S2/SvPasCrl or 129S1/SvImJ) mice by stereotaxic injection of elastase at the skull base, combined with systemic deoxycorticosterone acetate-salt hypertension. Neurological deficits and mortality were recorded. Aneurysms and subarachnoid hemorrhage grades were quantified postmortem, either after spontaneous mortality or at 7 to 21 days if the animals survived. In separate cohorts, we examined proinflammatory mediators by quantitative reverse transcriptase-polymerase chain reaction, arterial blood pressure via the femoral artery, and the circle of Willis by intravascular latex casting. RESULTS We found striking differences in aneurysm formation, rupture, and postrupture survival rates among the groups. 129Sv mice showed the highest rates of aneurysm rupture (80%), followed by C57 female (36%), C57 male (27%), and CD1 (21%). The risk of aneurysm rupture and the presence of unruptured aneurysms significantly differed among all 3 strains, as well as between male and female C57. The same hierarchy was observed upon Kaplan-Meier analysis of both overall survival and deficit-free survival. Subarachnoid hemorrhage grades were also more severe in 129Sv. CD1 mice showed the highest resistance to aneurysm rupture and the mildest outcomes. Higher mean blood pressures and the major phenotypic difference in the circle of Willis anatomy in 129Sv provided an explanation for the higher incidence of and more severe aneurysm ruptures. TNFα (tumor necrosis factor-alpha), IL-1β (interleukin-1-beta), and CCL2 (chemokine C-C motif ligand 2) expressions did not differ among the groups. CONCLUSIONS The outcome of elastase-induced intracranial aneurysm formation and rupture in mice depends on genetic background and shows sexual dimorphism.
Collapse
Affiliation(s)
- Takeshi Yanagisawa
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston. (T.Y., A.B.P.).,Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan (T.Y., Y.M.)
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill. (H.Z., J.E.F.)
| | - Tomoaki Suzuki
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.)
| | - Yoshinobu Kamio
- Department of Neurosurgery, Hamamatsu University School of Medicine, Japan (Y.K.)
| | - Tsubasa Takizawa
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.)
| | - Andreia Morais
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,National Institute of Translational Neuroscience, Biomedical Science Institute, Federal University of Rio de Janeiro, Brazil (A.M.)
| | - David Y Chung
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston. (D.Y.C., C.A.)
| | - Tao Qin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.)
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan (T.Y., Y.M.)
| | - James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill. (H.Z., J.E.F.).,Department of Neurobiology Curriculum, McAllister Heart Institute, University of North Carolina, Chapel Hill. (J.E.F.)
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston. (T.Y., A.B.P.)
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston. (D.Y.C., C.A.)
| |
Collapse
|
30
|
Abi-Ghanem C, Robison LS, Zuloaga KL. Androgens' effects on cerebrovascular function in health and disease. Biol Sex Differ 2020; 11:35. [PMID: 32605602 PMCID: PMC7328272 DOI: 10.1186/s13293-020-00309-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Androgens affect the cerebral vasculature and may contribute to sex differences in cerebrovascular diseases. Men are at a greater risk for stroke and vascular contributions to cognitive impairment and dementia (VCID) compared to women throughout much of the lifespan. The cerebral vasculature is a target for direct androgen actions, as it expresses several sex steroid receptors and metabolizing enzymes. Androgens’ actions on the cerebral vasculature are complex, as they have been shown to have both protective and detrimental effects, depending on factors such as age, dose, and disease state. When administered chronically, androgens are shown to be pro-angiogenic, promote vasoconstriction, and influence blood-brain barrier permeability. In addition to these direct effects of androgens on the cerebral vasculature, androgens also influence other vascular risk factors that may contribute to sex differences in cerebrovascular diseases. In men, low androgen levels have been linked to metabolic and cardiovascular diseases including hypertension, diabetes, hyperlipidemia, and obesity, which greatly increase the risk of stroke and VCID. Thus, a better understanding of androgens’ interactions with the cerebral vasculature under physiological and pathological conditions is of key importance.
Collapse
Affiliation(s)
- Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Lisa S Robison
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
31
|
Wiegers EJA, Mulder MJHL, Jansen IGH, Venema E, Compagne KCJ, Berkhemer OA, Emmer BJ, Marquering HA, van Es ACGM, Sprengers ME, van Zwam WH, van Oostenbrugge RJ, Roos YBWEM, Majoie CBLM, Roozenbeek B, Lingsma HF, Dippel DWJ, van der Lugt A. Clinical and Imaging Determinants of Collateral Status in Patients With Acute Ischemic Stroke in MR CLEAN Trial and Registry. Stroke 2020; 51:1493-1502. [PMID: 32279619 DOI: 10.1161/strokeaha.119.027483] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background and Purpose- Collateral circulation status at baseline is associated with functional outcome after ischemic stroke and effect of endovascular treatment. We aimed to identify clinical and imaging determinants that are associated with collateral grade on baseline computed tomography angiography in patients with acute ischemic stroke due to an anterior circulation large vessel occlusion. Methods- Patients included in the MR CLEAN trial (Multicenter Randomized Clinical Trial of Endovascular Treatment for Acute Ischemic Stroke in the Netherlands; n=500) and MR CLEAN Registry (n=1488) were studied. Collateral status on baseline computed tomography angiography was scored from 0 (absent) to 3 (good). Multivariable ordinal logistic regression analyses were used to test the association of selected determinants with collateral status. Results- In total, 1988 patients were analyzed. Distribution of the collateral status was as follows: absent (7%, n=123), poor (32%, n=596), moderate (39%, n=735), and good (23%, n=422). Associations for a poor collateral status in a multivariable model existed for age (adjusted common odds ratio, 0.92 per 10 years [95% CI, 0.886-0.98]), male (adjusted common odds ratio, 0.64 [95% CI, 0.53-0.76]), blood glucose level (adjusted common odds ratio, 0.97 [95% CI, 0.95-1.00]), and occlusion of the intracranial segment of the internal carotid artery with occlusion of the terminus (adjusted common odds ratio 0.50 [95% CI, 0.41-0.61]). In contrast to previous studies, we did not find an association between cardiovascular risk factors and collateral status. Conclusions- Older age, male sex, high glucose levels, and intracranial internal carotid artery with occlusion of the terminus occlusions are associated with poor computed tomography angiography collateral grades in patients with acute ischemic stroke eligible for endovascular treatment.
Collapse
Affiliation(s)
- Eveline J A Wiegers
- From the Department of Public Health (E.J.A.W., E.V., H.F.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maxim J H L Mulder
- Department of Neurology (M.J.H.L.M., E.V., K.C.J.C., O.A.B., B.R., D.W.J.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine (M.J.H.L.M., K.C.J.C., O.A.B., A.C.G.M.v.E., B.R., A.v.d.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ivo G H Jansen
- Department of Radiology and Nuclear Medicine (I.G.H.J., B.J.E., H.A.M., M.E.S., C.B.L.M.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Esmee Venema
- From the Department of Public Health (E.J.A.W., E.V., H.F.L.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Neurology (M.J.H.L.M., E.V., K.C.J.C., O.A.B., B.R., D.W.J.D.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Kars C J Compagne
- Department of Neurology (M.J.H.L.M., E.V., K.C.J.C., O.A.B., B.R., D.W.J.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine (M.J.H.L.M., K.C.J.C., O.A.B., A.C.G.M.v.E., B.R., A.v.d.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Olvert A Berkhemer
- Department of Neurology (M.J.H.L.M., E.V., K.C.J.C., O.A.B., B.R., D.W.J.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine (M.J.H.L.M., K.C.J.C., O.A.B., A.C.G.M.v.E., B.R., A.v.d.L.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Cardiovascular Research Institute Maastricht, the Netherlands (O.A.B., W.H.v.Z., R.J.v.O.)
| | - Bart J Emmer
- Department of Radiology and Nuclear Medicine (I.G.H.J., B.J.E., H.A.M., M.E.S., C.B.L.M.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Henk A Marquering
- Department of Radiology and Nuclear Medicine (I.G.H.J., B.J.E., H.A.M., M.E.S., C.B.L.M.M.), Amsterdam UMC, location AMC, the Netherlands.,Department of Biomedical Engineering and Physics (H.A.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Adriaan C G M van Es
- Department of Radiology and Nuclear Medicine (M.J.H.L.M., K.C.J.C., O.A.B., A.C.G.M.v.E., B.R., A.v.d.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marieke E Sprengers
- Department of Radiology and Nuclear Medicine (I.G.H.J., B.J.E., H.A.M., M.E.S., C.B.L.M.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Wim H van Zwam
- Cardiovascular Research Institute Maastricht, the Netherlands (O.A.B., W.H.v.Z., R.J.v.O.).,Department of Radiology (W.H.v.Z.), Maastricht University Medical Center, the Netherlands
| | - Robert J van Oostenbrugge
- Cardiovascular Research Institute Maastricht, the Netherlands (O.A.B., W.H.v.Z., R.J.v.O.).,Department of Neurology (R.J.v.O.), Maastricht University Medical Center, the Netherlands
| | - Yvo B W E M Roos
- Department of Neurology, Academic Medical Center, Amsterdam, the Netherlands (Y.B.W.E.M.R.)
| | - Charles B L M Majoie
- Department of Radiology and Nuclear Medicine (I.G.H.J., B.J.E., H.A.M., M.E.S., C.B.L.M.M.), Amsterdam UMC, location AMC, the Netherlands
| | - Bob Roozenbeek
- Department of Neurology (M.J.H.L.M., E.V., K.C.J.C., O.A.B., B.R., D.W.J.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Radiology and Nuclear Medicine (M.J.H.L.M., K.C.J.C., O.A.B., A.C.G.M.v.E., B.R., A.v.d.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Hester F Lingsma
- From the Department of Public Health (E.J.A.W., E.V., H.F.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Diederik W J Dippel
- Department of Neurology (M.J.H.L.M., E.V., K.C.J.C., O.A.B., B.R., D.W.J.D.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Aad van der Lugt
- Department of Radiology and Nuclear Medicine (M.J.H.L.M., K.C.J.C., O.A.B., A.C.G.M.v.E., B.R., A.v.d.L.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | | |
Collapse
|
32
|
León-Moreno LC, Castañeda-Arellano R, Rivas-Carrillo JD, Dueñas-Jiménez SH. Challenges and Improvements of Developing an Ischemia Mouse Model Through Bilateral Common Carotid Artery Occlusion. J Stroke Cerebrovasc Dis 2020; 29:104773. [PMID: 32199775 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104773] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/10/2020] [Accepted: 02/17/2020] [Indexed: 01/01/2023] Open
Abstract
Brain ischemia is one of the principal causes of death and disability worldwide in which prevention or an effective treatment does not exist. In order to develop successful treatments, an adequate and useful ischemia model is essential. Transient global cerebral ischemia is one of the most interesting pathological conditions in stroke studies because of the observed degeneration of forebrain and delayed neuronal cell death in selective vulnerable regions such as hippocampus. Transient occlusion of both common carotid arteries is the most convenient model to induce tGCI. Although there are effective rat and gerbil models using this method, the induction of a reproducible and reliable injury after global ischemia in mouse has presented higher variations, mainly because of its size and the necessary monitoring skills in order to accomplish homogeneous and reproducible results. Further, great variability among cerebral vasculature and susceptibility of the different strains and sub-strains is observed. In recent years, some modifications have been made to the model in order to normalize the heterogenic effects. Analysis of posterior communicating artery patency has been proposed as an exclusion parameter due to the direct relationship reported with the reduction of cerebral blood flow. Another method used to significantly reduce blood flow is the induction of hypotension with isoflurane. Each protocol produces distinct injury outcomes. Further improvements are needed to attain a general, simpler, reproducible and globally accepted model that allows comparisons between research groups, progress in understanding ischemia and the consequent development of therapeutic alternatives for ischemic injury.
Collapse
Affiliation(s)
| | - Rolando Castañeda-Arellano
- Department of Biomedical Sciences, University Center of Tonala, University de Guadalajara, Jalisco Mexico
| | - Jorge David Rivas-Carrillo
- Department of Physiology, Laboratory of Tissue Engineering and Transplant and cGMP Cell Processing Facility, Health Sciences Center, University de Guadalajara, Jalisco, Mexico
| | - Sergio Horacio Dueñas-Jiménez
- Department of Neuroscience, Laboratory of Neurophysiology, Health Sciences Center, University of Guadalajara, Guadalajara, C.P. 44340 Jalisco, México.
| |
Collapse
|
33
|
Foddis M, Winek K, Bentele K, Mueller S, Blumenau S, Reichhart N N, Crespo-Garcia S, Harnett D, Ivanov A, Meisel A, Joussen A, Strauss O, Beule D, Dirnagl U, Sassi C. An exploratory investigation of brain collateral circulation plasticity after cerebral ischemia in two experimental C57BL/6 mouse models. J Cereb Blood Flow Metab 2020; 40:276-287. [PMID: 31549895 PMCID: PMC7370619 DOI: 10.1177/0271678x19827251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Brain collateral circulation is an essential compensatory mechanism in response to acute brain ischemia. To study the temporal evolution of brain macro and microcollateral recruitment and their reciprocal interactions in response to different ischemic conditions, we applied a combination of complementary techniques (T2-weighted magnetic resonance imaging [MRI], time of flight [TOF] angiography [MRA], cerebral blood flow [CBF] imaging and histology) in two different mouse models. Hypoperfusion was either induced by permanent bilateral common carotid artery stenosis (BCCAS) or 60-min transient unilateral middle cerebral artery occlusion (MCAO). In both models, collateralization is a very dynamic phenomenon with a global effect affecting both hemispheres. Patency of ipsilateral posterior communicating artery (PcomA) represents the main variable survival mechanism and the main determinant of stroke lesion volume and recovery in MCAO, whereas the promptness of external carotid artery retrograde flow recruitment together with PcomA patency, critically influence survival, brain ischemic lesion volume and retinopathy in BCCAS mice. Finally, different ischemic gradients shape microcollateral density and size.
Collapse
Affiliation(s)
- Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katarzyna Winek
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kajetan Bentele
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Charité - Universitätsmedizin Berlin, NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadine Reichhart N
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sergio Crespo-Garcia
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dermot Harnett
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Andreas Meisel
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Antonia Joussen
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Olaf Strauss
- Department of Ophthalmology, Experimental Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health, BIH, Unit Bioinformatics, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,QUEST Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
34
|
Evaluation of sex differences in acid/base and electrolyte concentrations in acute large vessel stroke. Exp Neurol 2020; 323:113078. [DOI: 10.1016/j.expneurol.2019.113078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/26/2019] [Accepted: 10/01/2019] [Indexed: 12/26/2022]
|
35
|
Dula AN, Mlynash M, Zuck ND, Albers GW, Warach SJ. Neuroimaging in Ischemic Stroke Is Different Between Men and Women in the DEFUSE 3 Cohort. Stroke 2019; 51:481-488. [PMID: 31826731 DOI: 10.1161/strokeaha.119.028205] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Clinical deficits from ischemic stroke are more severe in women, but the pathophysiological basis of this sex difference is unknown. Sex differences in core and penumbral volumes and their relation to outcome were assessed in this substudy of the DEFUSE 3 clinical trial (Endovascular Therapy Following Imaging Evaluation for Ischemic Stroke). Methods- DEFUSE 3 randomized patients to thrombectomy or medical management who presented 6 to 16 hours from last known well with proximal middle cerebral artery or internal carotid artery occlusion and had target core and perfusion mismatch volumes on computed tomography or magnetic resonance imaging. Using univariate and adjusted regression models, the effect of sex was assessed on prerandomization measures of core, perfusion, and mismatch volumes and hypoperfusion intensity ratio, and on core volume growth using 24-hour scans. Results- All patients were included in the analysis (n=182) with 90 men and 92 women. There was no sex difference in the site of baseline arterial occlusion. Adjusted by age, baseline National Institutes of Health Stroke Scale, baseline modified Rankin Scale score, time to randomization, and imaging modality, women had smaller core, hypoperfusion, and penumbral volumes than men. Median (interquartile range) volumes for core were 8.0 mL (1.9-18.4) in women versus 12.6 mL (2.7-29.6) in men, for Tmax>6 seconds 89.0 mL (63.8-131.7) versus 133.9 mL (87.0-175.4), and for mismatch 82.1mL (53.8-112.8) versus 108.2 (64.1-149.2). The hypoperfusion intensity ratio was lower in women, 0.31 (0.15-0.46) versus 0.39 (0.26-0.57), P=0.006, indicating better collateral circulation, which was consistent with the observed slower ischemic core growth than men within the medical group (P=0.003). Conclusions- In the large vessel ischemic stroke cohort selected for DEFUSE 3, women had imaging evidence of better collateral circulation, smaller baseline core volumes, and slower ischemic core growth. These observations suggest sex differences in hemodynamic and temporal features of anterior circulation large artery occlusions. Registration- URL: https://www.clinicaltrials.gov. Unique identifier: NCT02586415.
Collapse
Affiliation(s)
- Adrienne N Dula
- From the Department of Neurology (A.N.D., N.D.Z., S.J.W.), Dell Medical School at The University of Texas, Austin
- Department of Diagnostic Medicine (A.N.D.), Dell Medical School at The University of Texas, Austin
| | - Michael Mlynash
- Stanford Stroke Center, Stanford University, Palo Alto, CA (M.M., G.W.A.)
| | - Nathan D Zuck
- From the Department of Neurology (A.N.D., N.D.Z., S.J.W.), Dell Medical School at The University of Texas, Austin
| | - Gregory W Albers
- Stanford Stroke Center, Stanford University, Palo Alto, CA (M.M., G.W.A.)
| | - Steven J Warach
- From the Department of Neurology (A.N.D., N.D.Z., S.J.W.), Dell Medical School at The University of Texas, Austin
| |
Collapse
|
36
|
Transient versus Permanent MCA Occlusion in Mice Genetically Modified to Have Good versus Poor Collaterals. ACTA ACUST UNITED AC 2019; 4. [PMID: 31840083 PMCID: PMC6910253 DOI: 10.20900/mo.20190024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Collateral-dependent blood flow is capable of significantly lessening the severity of stroke. Unfortunately, collateral flow varies widely in patients for reasons that remain unclear. Studies in mice have shown that the number and diameter of cerebral collaterals vary widely due primarily to polymorphisms in genes, e.g., Rabep2, involved in their formation during development. However, understanding how variation in collateral abundance affects stroke progression has been hampered by lack of a method to reversibly ligate the distal middle cerebral artery (MCAO) in mice. Here we present a method and examine infarct volume 24 h after transient (tMCAO, 90 min) versus permanent occlusion (pMCAO) in mice with good versus poor collaterals. Wildtype C57BL/6 mice (have abundant collaterals) sustained small infarctions following tMCAO that increased 2.1-fold after pMCAO, reflecting significant penumbra present at 90 min. Mutant C57BL/6 mice lacking Rabep2 (have reduced collaterals) sustained a 4-fold increase in infarct volume over WT following tMCAO and a smaller additional increase (0.4-fold) after pMCAO, reflecting reduced penumbra. Wildtype BALB/cBy (have a deficient Rabep2 variant and poor collaterals) had large infarctions following tMCAO that increased less (0.6-fold) than the above wildtype C57BL/6 mice following pMCAO. Mutant BALB/cBy mice (have deficient Rabep2 replaced with the C57BL/6 variant thus increased collaterals) sustained smaller infarctions after tMCAO. However, unlike C57BL/6 versus Rabep2 mice, penumbra was not increased since infarct volume increased only 0.3-fold following pMCAO. These findings present a murine model of tMCAO and demonstrate that neuroprotective mechanisms, in addition to collaterals, also vary with genetic background and affect the evolution of stroke.
Collapse
|
37
|
Quintana DD, Lewis SE, Anantula Y, Garcia JA, Sarkar SN, Cavendish JZ, Brown CM, Simpkins JW. The cerebral angiome: High resolution MicroCT imaging of the whole brain cerebrovasculature in female and male mice. Neuroimage 2019; 202:116109. [PMID: 31446129 PMCID: PMC6942880 DOI: 10.1016/j.neuroimage.2019.116109] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 01/09/2023] Open
Abstract
The cerebrovascular system provides crucial functions that maintain metabolic and homeostatic states of the brain. Despite its integral role of supporting cerebral viability, the topological organization of these networks remains largely uncharacterized. This void in our knowledge surmises entirely from current technological limitations that prevent the capturing of data through the entire depth of the brain. We report high-resolution reconstruction and analysis of the complete vascular network of the entire brain at the capillary level in adult female and male mice using a vascular corrosion cast procedure. Vascular network analysis of the whole brain revealed sex-related differences of vessel hierarchy. In addition, region-specific network analysis demonstrated different patterns of angioarchitecture between brain subregions and sex. Furthermore, our group is the first to provide a three-dimensional analysis of the angioarchitecture and network organization in a single reconstructed tomographic data set that encompasses all hierarchy of vessels in the brain of the adult mouse.
Collapse
Affiliation(s)
- D D Quintana
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - S E Lewis
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - Y Anantula
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J A Garcia
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - S N Sarkar
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J Z Cavendish
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - C M Brown
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J W Simpkins
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
38
|
Abstract
Cerebral small vessel disease (SVD) is characterized by changes in the pial and parenchymal microcirculations. SVD produces reductions in cerebral blood flow and impaired blood-brain barrier function, which are leading contributors to age-related reductions in brain health. End-organ effects are diverse, resulting in both cognitive and noncognitive deficits. Underlying phenotypes and mechanisms are multifactorial, with no specific treatments at this time. Despite consequences that are already considerable, the impact of SVD is predicted to increase substantially with the growing aging population. In the face of this health challenge, the basic biology, pathogenesis, and determinants of SVD are poorly defined. This review summarizes recent progress and concepts in this area, highlighting key findings and some major unanswered questions. We focus on phenotypes and mechanisms that underlie microvascular aging, the greatest risk factor for cerebrovascular disease and its subsequent effects.
Collapse
Affiliation(s)
- T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne Campus, Bundoora, Victoria 3086, Australia;
| | - Frank M Faraci
- Departments of Internal Medicine, Neuroscience, and Pharmacology, Francois M. Abboud Cardiovascular Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA;
| |
Collapse
|
39
|
Bonnin P, Mazighi M, Charriaut-Marlangue C, Kubis N. Early Collateral Recruitment After Stroke in Infants and Adults. Stroke 2019; 50:2604-2611. [DOI: 10.1161/strokeaha.119.025353] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Philippe Bonnin
- From the U965, INSERM, F-75010, Université de Paris, France (P.B.)
- U1148–Laboratory for Vascular and Translational Science, INSERM, F-75018, Université de Paris, France (P.B., M.M., N.K.)
- Service de Physiologie Clinique (P.B., N.K.), AP-HP, Hôpital Lariboisière, Paris, France
| | - Mikaël Mazighi
- U1148–Laboratory for Vascular and Translational Science, INSERM, F-75018, Université de Paris, France (P.B., M.M., N.K.)
- Service de Neurologie (M.M.), AP-HP, Hôpital Lariboisière, Paris, France
- Service de Neurologie, AP-HP, Hôpital Lariboisière, Paris, France (M.M.)
- Service de Neuroradiologie Interventionnelle, Fondation Rothschild, Paris, France (M.M.)
| | | | - Nathalie Kubis
- U1148–Laboratory for Vascular and Translational Science, INSERM, F-75018, Université de Paris, France (P.B., M.M., N.K.)
- Service de Physiologie Clinique (P.B., N.K.), AP-HP, Hôpital Lariboisière, Paris, France
| |
Collapse
|
40
|
Early Sex Differences in the Immune-Inflammatory Responses to Neonatal Ischemic Stroke. Int J Mol Sci 2019; 20:ijms20153809. [PMID: 31382688 PMCID: PMC6695584 DOI: 10.3390/ijms20153809] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
We recently reported that neonatal ischemia induces microglia/macrophage activation three days post-ischemia. We also found that female mice sustained smaller infarcts than males three months post-ischemia. The objective of our current study was to examine whether differential acute neuroinflammatory response and infiltrated immune cells occurs between male and females after three days post-ischemia. Permanent middle cerebral artery occlusion was induced in male and female postnatal 9-day-old (P9) mice, and mice were sacrificed three days after ischemia. Brains were analyzed for mRNA transcription after microglia magnetic cell sorting to evaluate M1 and M2 markers. FACS analysis was performed to assess myeloid infiltration and microglial expression of CX3 chemokine receptor 1 (CX3CR1). Inflammatory cytokine expression and microglia/macrophage activation were analyzed via in situ hybridization combined with immunofluorescence techniques. Lesion volume and cell death were measured. An increase in microglia/macrophages occurred in male versus female mice. The cells exhibited amoeboid morphology, and TNFα and ptgs2 (Cox-2) genes were more expressed in males. More myeloid cell infiltration was found in male versus female brains. However, we did not observe sex-dependent differences in the injured volume or cell death density. Our data show that sex differences in the acute microglial and immune responses to neonatal ischemia are likely both gene- and region-specific.
Collapse
|
41
|
Wang J, Lin X, Mu Z, Shen F, Zhang L, Xie Q, Tang Y, Wang Y, Zhang Z, Yang GY. Rapamycin Increases Collateral Circulation in Rodent Brain after Focal Ischemia as detected by Multiple Modality Dynamic Imaging. Am J Cancer Res 2019; 9:4923-4934. [PMID: 31410191 PMCID: PMC6691378 DOI: 10.7150/thno.32676] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/17/2019] [Indexed: 01/09/2023] Open
Abstract
Rationale: Brain collaterals contribute to improving ischemic stroke outcomes. However, dynamic and timely investigations of collateral blood flow and collateral restoration in whole brains of living animals have rarely been reported. Methods: Using multiple modalities of imaging, including synchrotron radiation angiography, laser speckle imaging, and micro-CT imaging, we dynamically explored collateral circulation throughout the whole brain in the rodent middle cerebral artery occlusion model. Results: We demonstrated that compared to control animals, 4 neocollaterals gradually formed between the intra- and extra-arteries in the skull base of model animals after occlusion (p<0.05). Two main collaterals were critical to the supply of blood from the posterior to the middle cerebral artery territory in the deep brain (p<0.05). Abundant small vessel and capillary anastomoses were detected on the surface of the cortex between the posterior and middle cerebral artery and between the anterior and middle cerebral artery (p<0.05). Collateral perfusion occurred immediately (≈15 min) and was maintained for up to 14 days after occlusion. Further study revealed that administration of rapamycin at 15 min after MCAO dilated the existing collateral vessels and promoted collateral perfusion. Principal conclusions: Our results provide evidence of collateral functional perfusion in the skull base, deep brain, and surface of the cortex. Rapamycin was capable of enlarging the diameter of collaterals, potentially extending the time window for ischemic stroke therapy.
Collapse
|
42
|
Li H, You W, Li X, Shen H, Chen G. Proteomic-Based Approaches for the Study of Ischemic Stroke. Transl Stroke Res 2019; 10:601-606. [PMID: 31278685 DOI: 10.1007/s12975-019-00716-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
43
|
Li P, Zhao G, Ding Y, Wang T, Flores J, Ocak U, Wu P, Zhang T, Mo J, Zhang JH, Tang J. Rh-IFN-α attenuates neuroinflammation and improves neurological function by inhibiting NF-κB through JAK1-STAT1/TRAF3 pathway in an experimental GMH rat model. Brain Behav Immun 2019; 79:174-185. [PMID: 30711510 PMCID: PMC6591046 DOI: 10.1016/j.bbi.2019.01.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/04/2019] [Accepted: 01/30/2019] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation occurs after germinal matrix hemorrhage (GMH) and induces secondary brain injury. Interferon-α (IFN-α) has been shown to exert anti-inflammatory effects in infectious diseases via activating IFNAR and its downstream signaling. We aimed to investigate the anti-inflammatory effects of Recombinant human IFN-α (rh-IFN-α) and the underlying mechanisms in a rat GMH model. Two hundred and eighteen P7 rat pups of both sexes were subjected to GMH by an intraparenchymal injection of bacterial collagenase. Rh-IFN-α was administered intraperitoneally. Small interfering RNA (siRNA) of IFNAR, and siRNA of tumor necrosis factor receptor associated factor 3 (TRAF3) were administered through intracerebroventricular (i.c.v.) injections. JAK1 inhibitor ruxolitinib was given by oral lavage. Post-GMH evaluation included neurobehavioral function, Nissl staining, Western blot analysis, and immunofluorescence. Our results showed that endogenous IFN-α and phosphorylated IFNAR levels were increased after GMH. Administration of rh-IFN-α improved neurological functions, attenuated neuroinflammation, inhibited microglial activation, and ameliorated post-hemorrhagic hydrocephalus after GMH. These observations were concomitant with IFNAR activation, increased expression of phosphorylated JAK1, phosphorylated STAT1 and TRAF3, and decreased levels of phosphorylated NF-κB, IL-6 and TNF-α. Specifically, knockdown of IFNAR, JAK1 and TRAF3 abolished the protective effects of rh-IFN-α. In conclusion, our findings demonstrated that rh-IFN-α treatment attenuated neuroinflammation, neurological deficits and hydrocephalus formation through inhibiting microglial activation after GMH, which might be mediated by IFNAR/JAK1-STAT1/TRAF3/NF-κB signaling pathway. Rh-IFN-α may be a promising therapeutic agent to attenuate brain injury via its anti-inflammatory effect.
Collapse
Affiliation(s)
- Peng Li
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China; Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou 510180, China
| | - Gang Zhao
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Department of Emergency Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China; Traumatic Research Center of Yunnan Province, Kunming 650101, China
| | - Yan Ding
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Tianyi Wang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Jerry Flores
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Umut Ocak
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Pei Wu
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Tongyu Zhang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - Jun Mo
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States; Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Science, School of Medicine, Loma Linda University, Loma Linda, CA 92354, United States.
| |
Collapse
|
44
|
Ding Y, Flores J, Klebe D, Li P, McBride DW, Tang J, Zhang JH. Annexin A1 attenuates neuroinflammation through FPR2/p38/COX-2 pathway after intracerebral hemorrhage in male mice. J Neurosci Res 2019; 98:168-178. [PMID: 31157469 DOI: 10.1002/jnr.24478] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 04/08/2019] [Accepted: 05/15/2019] [Indexed: 11/08/2022]
Abstract
Spontaneous intracerebral hemorrhage (ICH) is the deadliest stroke subtype and neuroinflammation is a critical component of the pathogenesis following ICH. Annexin A1-FPR2 signaling has been shown to play a protective role in animal stroke models. This study aimed to assess whether Annexin A1 attenuated neuroinflammation and brain edema after ICH and investigate the underlying mechanisms. Male CD-1 mice were subjected to collagenase-induced ICH. Annexin A1 was administered at 0.5 hr after ICH. Brain water content measurement, short-term and long-term neurobehavioral tests, Western blot and immnunofluorescence were performed. Results showed that Annexin A1 effectively attenuated brain edema, improved short-term neurological function and ameliorated microglia activation after ICH. Annexin A1 also improved memory function at 28 days after ICH. However, these beneficial effects were abolished with the administration of FPR2 antagonist Boc-2. Furthermore, AnxA1/FPR2 signaling may confer protective effects via inhibiting p38-associated inflammatory cascade. Our study demonstrated that Annexin A1/FPR2/p38 signaling pathway played an important role in attenuating neuroinflammation after ICH and that Annexin A1 could be a potential therapeutic strategy for ICH patients.
Collapse
Affiliation(s)
- Yan Ding
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jerry Flores
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - Damon Klebe
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - Peng Li
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Jiping Tang
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California.,Departments of Anesthesiology, Neurology and Neurosurgery, Loma Linda University, Loma Linda, California
| |
Collapse
|
45
|
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
|
46
|
Zhao L, Mulligan MK, Nowak TS. Substrain- and sex-dependent differences in stroke vulnerability in C57BL/6 mice. J Cereb Blood Flow Metab 2019; 39:426-438. [PMID: 29260927 PMCID: PMC6421252 DOI: 10.1177/0271678x17746174] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The C57BL/6 mouse strain is represented by distinct substrains, increasingly recognized to differ genetically and phenotypically. The current study compared stroke vulnerability among C57BL/6 J (J), C57BL/6JEiJ (JEiJ), C57BL/6ByJ (ByJ), C57BL/6NCrl (NCrl), C57BL/6NJ (NJ) and C57BL/6NTac (NTac) substrains, using a model of permanent distal middle cerebral artery and common carotid artery occlusion. Mean infarct volume was nearly two-fold smaller in J, JEiJ and ByJ substrains relative to NCrl, NJ and NTac (N-lineage) mice. This identifies a previously unrecognized confound in stroke studies involving genetically modified strain comparisons if control substrain background were not rigorously matched. Mean infarct size was smaller in females of J and ByJ substrains than in the corresponding males, but there was no sex difference for NCrl and NJ mice. A higher proportion of small infarcts in J and ByJ substrains was largely responsible for both substrain- and sex-dependent differences. These could not be straightforwardly explained by variations in posterior communicating artery patency, MCA anatomy or acute penumbral blood flow deficits. Their larger and more homogeneously distributed infarcts, together with their established use as the common background for many genetically modified strains, may make N-lineage C57BL/6 substrains the preferred choice for future studies in experimental stroke.
Collapse
Affiliation(s)
- Liang Zhao
- 1 Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Megan K Mulligan
- 2 Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Thaddeus S Nowak
- 1 Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
47
|
Mouse models of Alzheimer's disease cause rarefaction of pial collaterals and increased severity of ischemic stroke. Angiogenesis 2019; 22:263-279. [PMID: 30519973 PMCID: PMC6475514 DOI: 10.1007/s10456-018-9655-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 11/20/2018] [Indexed: 01/26/2023]
Abstract
Vascular dysfunction contributes to the progression and severity of Alzheimer's disease (AD). Patients with AD also sustain larger infarctions after ischemic stroke; however, the responsible mechanisms are unknown. Pial collaterals are the primary source of protection in stroke. Unfortunately, natural aging and other vascular risk factors cause a decline in collateral number and diameter (rarefaction) and an increase in stroke severity. Herein, we tested the hypothesis that AD accelerates age-induced collateral rarefaction and examined potential underlying mechanisms. Triple and double transgenic mouse models of AD both sustained collateral rarefaction by 8 months of age, well before the onset of rarefaction caused by aging alone (16 months of age). Rarefaction, which did not progress further at 18 months of age, was accompanied by a twofold increase in infarct volume after MCA occlusion. AD did not induce rarefaction of similarly sized pial arterioles or penetrating arterioles. Rarefaction was minimal and occurred only at 18 months of age in a parenchymal vascular amyloid-beta model of AD. Rarefaction was not associated with amyloid-beta deposition on collaterals or pial arteries, nor was plaque burden or CD11b+ cell density greater in brain underlying the collateral zones versus elsewhere. However, rarefaction was accompanied by increased markers of oxidative stress, inflammation, and aging of collateral endothelial and mural cells. Moreover, rarefaction was lessened by deletion of CX3CR1 and prevented by overexpression of eNOS. These findings demonstrate that mouse models of AD promote rarefaction of pial collaterals and implicate inflammation-induced accelerated aging of collateral wall cells. Strategies that reduce vascular inflammation and/or increase nitric oxide may preserve collateral function.
Collapse
|
48
|
Robison LS, Gannon OJ, Salinero AE, Zuloaga KL. Contributions of sex to cerebrovascular function and pathology. Brain Res 2018; 1710:43-60. [PMID: 30580011 DOI: 10.1016/j.brainres.2018.12.030] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
Sex differences exist in how cerebral blood vessels function under both physiological and pathological conditions, contributing to observed sex differences in risk and outcomes of cerebrovascular diseases (CBVDs), such as vascular contributions to cognitive impairment and dementia (VCID) and stroke. Throughout most of the lifespan, women are protected from CBVDs; however, risk increases following menopause, suggesting sex hormones may play a significant role in this protection. The cerebrovasculature is a target for sex hormones, including estrogens, progestins, and androgens, where they can influence numerous vascular functions and pathologies. While there is a plethora of information on estrogen, the effects of progestins and androgens on the cerebrovasculature are less well-defined. Estrogen decreases cerebral tone and increases cerebral blood flow, while androgens increase tone. Both estrogens and androgens enhance angiogenesis/cerebrovascular remodeling. While both estrogens and androgens attenuate cerebrovascular inflammation, pro-inflammatory effects of androgens under physiological conditions have also been demonstrated. Sex hormones exert additional neuroprotective effects by attenuating oxidative stress and maintaining integrity and function of the blood brain barrier. Most animal studies utilize young, healthy, gonadectomized animals, which do not mimic the clinical conditions of aging individuals likely to get CBVDs. This is also concerning, as sex hormones appear to mediate cerebrovascular function differently based on age and disease state (e.g. metabolic syndrome). Through this review, we hope to inspire others to consider sex as a key biological variable in cerebrovascular research, as greater understanding of sex differences in cerebrovascular function will assist in developing personalized approaches to prevent and treat CBVDs.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| |
Collapse
|
49
|
Gannon OJ, Robison LS, Custozzo AJ, Zuloaga KL. Sex differences in risk factors for vascular contributions to cognitive impairment & dementia. Neurochem Int 2018; 127:38-55. [PMID: 30471324 DOI: 10.1016/j.neuint.2018.11.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/11/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is the second most common cause of dementia. While males overall appear to be at a slightly higher risk for VCID throughout most of the lifespan (up to age 85), some risk factors for VCID more adversely affect women. These include female-specific risk factors associated with pregnancy related disorders (e.g. preeclampsia), menopause, and poorly timed hormone replacement. Further, presence of certain co-morbid risk factors, such as diabetes, obesity and hypertension, also may more adversely affect women than men. In contrast, some risk factors more greatly affect men, such as hyperlipidemia, myocardial infarction, and heart disease. Further, stroke, one of the leading risk factors for VCID, has a higher incidence in men than in women throughout much of the lifespan, though this trend is reversed at advanced ages. This review will highlight the need to take biological sex and common co-morbidities for VCID into account in both preclinical and clinical research. Given that there are currently no treatments available for VCID, it is critical that we understand how to mitigate risk factors for this devastating disease in both sexes.
Collapse
Affiliation(s)
- O J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| | - L S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| | - A J Custozzo
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| | - K L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| |
Collapse
|
50
|
Li Z, Tremble SM, Cipolla MJ. Implications for understanding ischemic stroke as a sexually dimorphic disease: the role of pial collateral circulations. Am J Physiol Heart Circ Physiol 2018; 315:H1703-H1712. [PMID: 30239233 DOI: 10.1152/ajpheart.00402.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We investigated structural and functional differences in primary and pial collateral circulations in adult normotensive male and female Wistar rats. Male ( n = 10) and female ( n = 7) rats were subjected to middle cerebral artery (MCA) occlusion and changes in relative cerebral blood flow in MCA and pial collateral territories were measured by multisite laser-Doppler flowmetry. Rats were then transcardially perfused with a mixture of carbon black and latex, perfusion fixed, and imaged to compare primary and pial collateral structure between male ( n = 4) and female ( n = 3) rats, including lumen diameters and number. To study pial collateral function, leptomeningeal anastomoses (LMAs) were isolated and pressurized from male ( n = 7) and female ( n = 6) rats. Myogenic tone and reactivity to pressure, vascular function to pharmacological activator, or inhibitor of ion channels was measured and compared. There was no difference between relative cerebral blood flow in both MCA and pial collateral territories during occlusion and reperfusion between groups. Compared with male LMAs, female LMAs had similar myogenic tone (24.0 ± 7.3% vs. 16.0 ± 3.7%, P > 0.05) and reactivity to increased pressure and similar vascular responses to vasoconstrictive and vasodilatory stimuli. Additionally, compared with female LMAs, male LMAs had similar numbers (21 ± 1 vs. 20 ± 2, P > 0.05) and diameters (30.5 ± 2.0 vs. 26.2 ± 0.6 μm, P > 0.05), and no sex difference was detected in the diameter of arterial segments of circle of Willis. Together, our data establish no sex difference of cerebral collateral structure or function, suggesting that the reduced severity of stroke outcome in female rats is not likely due to differences in the cerebral collateral circulation. NEW & NOTEWORTHY Our work compared the function of leptomeningeal anastomoses between male and female adult normotensive rats with no sex difference found. We also confirmed no sex difference in primary and pial collateral structure in Wistar rats. Our findings suggest that the reduced severity of stroke in premenopausal women and reproductively intact female rodents is not likely due to improved primary and pial collateral circulations.
Collapse
Affiliation(s)
- Zhaojin Li
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont
| | - Sarah M Tremble
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont
| | - Marilyn J Cipolla
- Department of Neurological Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont.,Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Robert Larner College of Medicine , Burlington, Vermont.,Department of Pharmacology, University of Vermont Robert Larner College of Medicine , Burlington, Vermont
| |
Collapse
|