1
|
Choi YH, Hsu M, Laaker C, Port J, Kovács KG, Herbath M, Yang H, Cismaru P, Johnson AM, Spellman B, Wigand K, Sandor M, Fabry Z. Dual role of vascular endothelial growth factor-C in post-stroke recovery. J Exp Med 2025; 222:e20231816. [PMID: 39665829 PMCID: PMC11636551 DOI: 10.1084/jem.20231816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 09/25/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Cerebrospinal fluid (CSF), antigens, and antigen-presenting cells drain from the central nervous system (CNS) into lymphatic vessels near the cribriform plate and dura, yet the role of these vessels during stroke is unclear. Using a mouse model of ischemic stroke, transient middle cerebral artery occlusion (tMCAO), we demonstrate stroke-induced lymphangiogenesis near the cribriform plate, peaking at day 7 and regressing by day 14. Lymphangiogenesis is restricted to the cribriform plate and deep cervical lymph nodes and is regulated by VEGF-C/VEGFR-3 signaling. The use of a VEGFR-3 inhibitor prevented lymphangiogenesis and led to improved stroke outcomes at earlier time points, with no effects at later time points. VEGF-C delivery after tMCAO did not further increase post-stroke lymphangiogenesis, but instead induced larger brain infarcts. Our data support the damaging role of VEGF-C acutely and a pro-angiogenic role chronically. This nuanced understanding of VEGFR-3 and VEGF-C in stroke pathology advises caution regarding therapeutic VEGF-C use in stroke.
Collapse
Affiliation(s)
- Yun Hwa Choi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Martin Hsu
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, Chapel Hill, NC, USA
| | - Collin Laaker
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Jenna Port
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristóf G. Kovács
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Melinda Herbath
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Heeyoon Yang
- College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter Cismaru
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Alexis M. Johnson
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Bailey Spellman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Kelsey Wigand
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Jia J, Niu L, Feng P, Liu S, Han H, Zhang B, Wang Y, Wang M. Identification of Novel Biomarkers for Ischemic Stroke Through Integrated Bioinformatics Analysis and Machine Learning. J Mol Neurosci 2025; 75:13. [PMID: 39862324 DOI: 10.1007/s12031-025-02309-8] [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: 12/04/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Ischemic stroke leads to permanent damage to the affected brain tissue, with strict time constraints for effective treatment. Predictive biomarkers demonstrate great potential in the clinical diagnosis of ischemic stroke, significantly enhancing the accuracy of early identification, thereby enabling clinicians to intervene promptly and reduce patient disability and mortality rates. Furthermore, the application of predictive biomarkers facilitates the development of personalized treatment plans tailored to the specific conditions of individual patients, optimizing treatment outcomes and improving prognoses. Bioinformatics technologies based on high-throughput data provide a crucial foundation for comprehensively understanding the biological characteristics of ischemic stroke and discovering effective predictive targets. In this study, we evaluated gene expression data from ischemic stroke patients retrieved from the Gene Expression Omnibus (GEO) database, conducting differential expression analysis and functional analysis. Through weighted gene co-expression network analysis (WGCNA), we characterized gene modules associated with ischemic stroke. To screen candidate core genes, three machine learning algorithms were applied, including Least Absolute Shrinkage and Selection Operator (LASSO), random forest (RF), and support vector machine-recursive feature elimination (SVM-RFE), ultimately identifying five candidate core genes: MBOAT2, CKAP4, FAF1, CLEC4D, and VIM. Subsequent validation was performed using an external dataset. Additionally, the immune infiltration landscape of ischemic stroke was mapped using the CIBERSORT method, investigating the relationship between candidate core genes and immune cells in the pathogenesis of ischemic stroke, as well as the key pathways associated with the core genes. Finally, the key gene VIM was further identified and preliminarily validated through four machine learning algorithms, including generalized linear model (GLM), Extreme Gradient Boosting (XGBoost), RF, and SVM-RFE. This study contributes to advancing our understanding of biomarkers for ischemic stroke and provides a reference for the prediction and diagnosis of ischemic stroke.
Collapse
Affiliation(s)
- Juan Jia
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China
- Department of Anesthesiology, Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Liang Niu
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Peng Feng
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China
| | - Shangyu Liu
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China
| | - Hongxi Han
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China
| | - Bo Zhang
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China
| | - Yingbin Wang
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China.
- Department of Anesthesiology, Second Hospital of Lanzhou University, Lanzhou, 730030, China.
| | - Manxia Wang
- Lanzhou University Second Hospital, The Second Medical College of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China.
- Department of Neurology, Second Hospital of Lanzhou University, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, China.
| |
Collapse
|
3
|
Kozler P, Marešová D, Pokorný J. Assessment of Blood-Brain Barrier Permeability in a Cerebral Ischemia-Reperfusion Model in Rats; A Pilot Study. Physiol Res 2024; 73:1099-1105. [PMID: 39903898 PMCID: PMC11835218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/12/2024] [Indexed: 02/06/2025] Open
Abstract
Animal models are an important tool for studying ischemic mechanisms of stroke. Among them, the middle cerebral artery occlusion (MCAO) model via the intraluminal suture method in rodents is closest to human ischemic stroke. It is a model of transient occlusion followed by reperfusion, thus representing cerebral ischemia-reperfusion model that simulates patients with vascular occlusion and timely recanalization. Although reperfusion is very beneficial for the possibility of preserving brain functions after ischemia, it also brings a great risk in the form of brain edema, which can cause the development of intracranial hypertension, and increasing morbidity and mortality. In this paper, we present the results of our own transient reperfusion model of MCAO in which we tested the permeability of the blood-brain barrier (BBB) using Evans blue (EB), an intravital dye with a high molecular weight (68,500 Da) that prevents its penetration through the intact BBB. A total of 15 animals were used in the experiment and underwent the following procedures: insertion of the MCA occluder; assessment of ischemia by 2,3,5 -Triphenyltetrazolium chloride (TTC) staining; assessment of the BBB permeability using brain EB distribution. The results are presented and discussed. The test of BBB permeability using EB showed that 120 minutes after induction of ischemia, the BBB is open for the entry of large molecules into the brain. We intend to use this finding to time the application of neuroprotective agents via ICA injection in our next stroke model. Keywords: Cerebral ischemia-reperfusion model, Middle cerebral artery occlusion, Blood-brain barrier, 2,3,5 -Triphenyltetrazolium chloride, Evans blue.
Collapse
Affiliation(s)
- P Kozler
- Institute of Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | |
Collapse
|
4
|
Komatsu T, Ohta H, Takeda M, Matsumura Y, Yokoyama M, Wang Z, Okano HJ, Iguchi Y. Novel Rat Model of Embolic Cerebral Ischemia Using a Radiopaque Blood Clot and a Microcatheter Under Fluoroscopy. Transl Stroke Res 2024:10.1007/s12975-024-01312-2. [PMID: 39560687 DOI: 10.1007/s12975-024-01312-2] [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: 08/23/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Conventional rat models of thromboembolic stroke do not allow control of infarct size or spontaneous recanalization. We aimed to develop a novel rat thromboembolic stroke model that ensures highly reproducible infarct sizes and locations within the MCA territory and does not require arterial ligation. Twenty male Sprague-Dawley rats and two sham-operated rats were included. A microcatheter was navigated from the caudal ventral artery to the internal carotid artery using digital subtraction angiography. A blood clot (diameter, 0.86 mm; length, 3 mm) containing zirconium dioxide was advanced in the catheter. Fluoroscopy was performed at 1, 3, 6, and 24 h after stroke model creation, and TTC staining was conducted at 24 h. Neurological deficit scores were measured. In all embolized rats, the ACA and MCA bifurcation were selective. Median operating time was 6 min. The position of the radiopaque blood clot remained unchanged for 24 h after model creation in 19/20 rats. Median infarct volume was 242 mm3 (IQR, 239-262 mm3). We present a novel rat model of highly reproducible focal infarct in only the MCA territory. Fluoroscopy effectively identified any blood clot migration. This model could contribute to the development of new thrombolytic agents.
Collapse
Affiliation(s)
- Teppei Komatsu
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| | - Hiroki Ohta
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Misato Takeda
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-Ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Masayuki Yokoyama
- Division of Medical Engineering, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Zuojun Wang
- Division of Artificial Intelligence in Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| |
Collapse
|
5
|
Hu X, Han M, Liu J, Li F, Cui Y. Ameliorative Effect of Natural Sesquiterpene Alcohol Cedrol Against Cerebral Ischemia Infarction-In Vitro and In Vivo Studies. Appl Biochem Biotechnol 2024; 196:8026-8042. [PMID: 38668841 DOI: 10.1007/s12010-024-04965-9] [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] [Accepted: 04/16/2024] [Indexed: 12/14/2024]
Abstract
Cedrol is a major bioactive compound present in the Cedrus atlantica with numerous biological properties. In this study, we elucidated the neuroprotective properties of cedrol against ischemic infarction in animal and in vitro studies. A cerebral ischemic/reperfusion model was induced in adult Wistar rats, and oxygen-glucose deprivation/reperfusion was induced in SH-SY5Y neuronal cells and treated with different concentrations of cedrol. The percentage of water content, cerebral infarct, and neurological deficit score was assessed in experimental rats. The acetylcholinesterase activity and inflammatory cytokines were quantified to analyze the anti-inflammatory potency of cedrol. Oxidative stress marker malondialdehyde and antioxidants were quantified to evaluate the antioxidant potency of cedrol in an ischemic condition. The neuroprotective potency of cedrol was confirmed by histopathological analysis of the brain tissue of cedrol-treated I/R-induced rats. In in vitro studies, the MTT and LDH assays were performed in cedrol-treated OGD/R SH-SY5Y cells to analyze the cytoprotective effect of cedrol. The anti-inflammatory property of cedrol was confirmed by quantifying the pro-inflammatory cytokine levels in OGD/R-induced cedrol-treated SH-SY5Y cells. The results obtained prove that cedrol significantly prevents brain edema, neurological deficits, acetylcholinesterase activity, and oxidative damage in ischemic-induced rats. It inhibited neuroinflammation in ischemic-induced rats and also in in vitro models. The neuroprotective effect of cedrol during an ischemic condition was authentically established with histological analysis in an animal model and cell survival assays in an in vitro model. Overall, our results confirm that cedrol is a potent alternative drug to treat cerebral ischemia in the future.
Collapse
Affiliation(s)
- Xiaohong Hu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mei Han
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Feng Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanchao Cui
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
6
|
Li J, Liu X, Wang Z, Xie P, Zhu M, Zhong H, Luo S, Tang J, Mo G. Ozone therapy mitigates parthanatos after ischemic stroke. Biol Res 2024; 57:71. [PMID: 39367424 PMCID: PMC11453019 DOI: 10.1186/s40659-024-00547-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/05/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Stroke is a leading cause of death worldwide, with oxidative stress and calcium overload playing significant roles in the pathophysiology of the disease. Ozone, renowned for its potent antioxidant properties, is commonly employed as an adjuvant therapy in clinical settings. Nevertheless, it remains unclear whether ozone therapy on parthanatos in cerebral ischemia-reperfusion injury (CIRI). This study aims to investigate the impact of ozone therapy on reducing parthanatos during CIRI and to elucidate the underlying mechanism. METHODS Hydrogen peroxide (H2O2) was utilized to mimic the generation of reactive oxygen species (ROS) in SH-SY5Y cell reperfusion injury in vitro, and an in vivo ischemic stroke model was established. Ozone saline was introduced for co-culture or intravenously administered to mice. Apoptosis and oxidative stress were assessed using flow cytometry and immunofluorescence. Western blotting was utilized to examine the expression of parthanatos signature proteins. The mechanism by which ozone inhibits parthanatos was elucidated through inhibiting PPARg or Nrf2 activity. RESULTS The findings demonstrated that ozone mitigated H2O2-induced parthanatos by either upregulating nuclear factor erythroid 2-related factor 2 (Nrf2) or activating peroxisome proliferator-activated receptorg (PPARg). Furthermore, through the use of calcium chelators and ROS inhibitors, it was discovered that ROS directly induced parthanatos and facilitated intracellular calcium elevation. Notably, a malignant feedback loop between ROS and calcium was identified, further amplifying the induction of parthanatos. Ozone therapy exhibited its efficacy by increasing PPARg activity or enhancing the Nrf2 translation, thereby inhibiting ROS production induced by H2O2. Concurrently, our study demonstrated that ozone treatment markedly inhibited parthanatos in stroke-afflicted mice. Additionally, ozone therapy demonstrated significant neuroprotective effects on cortical neurons, effectively suppressing parthanatos. CONCLUSIONS These findings contribute valuable insights into the potential of ozone therapy as a therapeutic strategy for reducing parthanatos during CIRI, highlighting its impact on key molecular pathways associated with oxidative stress and calcium regulation.
Collapse
Affiliation(s)
- Jiahui Li
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xiaolei Liu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zengze Wang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Pengyun Xie
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Min Zhu
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Hanhui Zhong
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Sirui Luo
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jing Tang
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Guixi Mo
- The Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
7
|
Mușat MI, Cătălin B, Hadjiargyrou M, Popa-Wagner A, Greșiță A. Advancing Post-Stroke Depression Research: Insights from Murine Models and Behavioral Analyses. Life (Basel) 2024; 14:1110. [PMID: 39337894 PMCID: PMC11433193 DOI: 10.3390/life14091110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Post-stroke depression (PSD) represents a significant neuropsychiatric complication that affects between 39% and 52% of stroke survivors, leading to impaired recovery, decreased quality of life, and increased mortality. This comprehensive review synthesizes our current knowledge of PSD, encompassing its epidemiology, risk factors, underlying neurochemical mechanisms, and the existing tools for preclinical investigation, including animal models and behavioral analyses. Despite the high prevalence and severe impact of PSD, challenges persist in accurately modeling its complex symptomatology in preclinical settings, underscoring the need for robust and valid animal models to better understand and treat PSD. This review also highlights the multidimensional nature of PSD, where both biological and psychosocial factors interplay to influence its onset and course. Further, we examine the efficacy and limitations of the current animal models in mimicking the human PSD condition, along with behavioral tests used to evaluate depressive-like behaviors in rodents. This review also sets a new precedent by integrating the latest findings across multidisciplinary studies, thereby offering a unique and comprehensive perspective of existing knowledge. Finally, the development of more sophisticated models that closely replicate the clinical features of PSD is crucial in order to advance translational research and facilitate the discovery of future effective therapies.
Collapse
Affiliation(s)
- Mădălina Iuliana Mușat
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bogdan Cătălin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Aurel Popa-Wagner
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Neurology, Vascular Neurology and Dementia, University of Medicine Essen, 45122 Essen, Germany
| | - Andrei Greșiță
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, NY 11568, USA
| |
Collapse
|
8
|
Wang J, Li Y, Qi L, Mamtilahun M, Liu C, Liu Z, Shi R, Wu S, Yang GY. Advanced rehabilitation in ischaemic stroke research. Stroke Vasc Neurol 2024; 9:328-343. [PMID: 37788912 PMCID: PMC11420926 DOI: 10.1136/svn-2022-002285] [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: 12/30/2022] [Accepted: 03/20/2023] [Indexed: 10/05/2023] Open
Abstract
At present, due to the rapid progress of treatment technology in the acute phase of ischaemic stroke, the mortality of patients has been greatly reduced but the number of disabled survivors is increasing, and most of them are elderly patients. Physicians and rehabilitation therapists pay attention to develop all kinds of therapist techniques including physical therapy techniques, robot-assisted technology and artificial intelligence technology, and study the molecular, cellular or synergistic mechanisms of rehabilitation therapies to promote the effect of rehabilitation therapy. Here, we discussed different animal and in vitro models of ischaemic stroke for rehabilitation studies; the compound concept and technology of neurological rehabilitation; all kinds of biological mechanisms of physical therapy; the significance, assessment and efficacy of neurological rehabilitation; the application of brain-computer interface, rehabilitation robotic and non-invasive brain stimulation technology in stroke rehabilitation.
Collapse
Affiliation(s)
- Jixian Wang
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medical, Shanghai, China
| | - Yongfang Li
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medical, Shanghai, China
| | - Lin Qi
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Muyassar Mamtilahun
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Liu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ze Liu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Rubing Shi
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shengju Wu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Komatsu T, Ohta H, Takakura N, Hata J, Kitagawa T, Kurashina Y, Onoe H, Okano HJ, Iguchi Y. A Novel Rat Model of Embolic Cerebral Ischemia Using a Cell-Implantable Radiopaque Hydrogel Microfiber. Transl Stroke Res 2024; 15:636-646. [PMID: 36867349 DOI: 10.1007/s12975-023-01144-6] [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: 01/20/2023] [Revised: 02/12/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
The failure of neuroprotective treatment-related clinical trials, including stem cell therapies, may be partially due to a lack of suitable animal models. We have developed a stem cell-implantable radiopaque hydrogel microfiber that can survive for a long time in vivo. The microfiber is made of barium alginate hydrogel containing zirconium dioxide, fabricated in a dual coaxial laminar flow microfluidic device. We aimed to develop a novel focal stroke model using this microfiber. Using male Sprague-Dawley rats (n=14), a catheter (inner diameter, 0.42 mm; outer diameter, 0.55 mm) was navigated from the caudal ventral artery to the left internal carotid artery using digital subtraction angiography. A radiopaque hydrogel microfiber (diameter, 0.4 mm; length, 1 mm) was advanced through the catheter by slow injection of heparinized physiological saline to establish local occlusion. Both 9.4-T magnetic resonance imaging at 3 and 6 h and 2% 2,3,5-triphenyl tetrazolium chloride staining at 24 h after stroke model creation were performed. Neurological deficit score and body temperature were measured. The anterior cerebral artery-middle cerebral artery bifurcation was selectively embolized in all rats. Median operating time was 4 min (interquartile range [IQR], 3-8 min). Mean infarct volume was 388 mm3 (IQR, 354-420 mm3) at 24 h after occlusion. No infarction of the thalamus or hypothalamus was seen. Body temperature did not change significantly over time (P = 0.204). However, neurological deficit scores before and at 3, 6, and 24 h after model creation differed significantly (P < 0.001). We present a novel rat model of focal infarct restricted to the middle cerebral artery territory using a radiopaque hydrogel microfiber positioned under fluoroscopic guidance. By comparing the use of stem cell-containing versus non-containing fibers in this stroke model, it would be possible to determine the efficacy of "pure" cell transplantation in treating stroke.
Collapse
Affiliation(s)
- Teppei Komatsu
- Department of Neurology, the Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-ku, Tokyo, Japan, 105-8461.
| | - Hiroki Ohta
- Division of Regenerative Medicine, Research Center for Medical Sciences, the Jikei University School of Medicine, Tokyo, Japan
| | - Naoki Takakura
- School of integrated DESIGN Engineering, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Junichi Hata
- Department of Radiological Science, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Tomomichi Kitagawa
- Department of Neurology, the Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-ku, Tokyo, Japan, 105-8461
| | - Yuta Kurashina
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, Kanagawa, Japan
- Division of Advanced Mechanical Systems Engineering, Institute of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, the Jikei University School of Medicine, Tokyo, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, the Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-ku, Tokyo, Japan, 105-8461
| |
Collapse
|
10
|
Raza SS. Rat Model of Middle Cerebral Artery Occlusion. Methods Mol Biol 2024; 2761:623-633. [PMID: 38427265 DOI: 10.1007/978-1-0716-3662-6_41] [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] [Indexed: 03/02/2024]
Abstract
Stroke is the third-leading cause of death and the leading cause of acquired adult disability worldwide. Several ischemic stroke models are currently available. However, mimicking focal cerebral ischemia (FCI) is the most common. The formation of an embolic or thrombotic occlusion at or near the middle cerebral artery causes most events in FCI. The current protocol closely mimics the etiology of human stroke and ensures that the results obtained are highly relevant. The method described in this protocol yields reproducible results. The success of this model in ischemic research can be examined through the utilization of Doppler blood flow imaging equipment.
Collapse
Affiliation(s)
- Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India.
- Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Lucknow, India.
| |
Collapse
|
11
|
Sethiya NK, Ghiloria N, Srivastav A, Bisht D, Chaudhary SK, Walia V, Alam MS. Therapeutic Potential of Myricetin in the Treatment of Neurological, Neuropsychiatric, and Neurodegenerative Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:865-882. [PMID: 37461364 DOI: 10.2174/1871527322666230718105358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 06/12/2024]
Abstract
Myricetin (MC), 3,5,7,3',4',5'-hexahydroxyflavone, chemically belongs to a flavonoid category known to confer antioxidant, antimicrobial, antidiabetic, and neuroprotective effects. MC is known to suppress the generation of Reactive Oxygen Species (ROS), lipid peroxidation (MDA), and inflammatory markers. It has been reported to improve insulin function in the human brain and periphery. Besides this, it modulates several neurochemicals including glutamate, GABA, serotonin, etc. MC has been shown to reduce the expression of the enzyme Mono Amine Oxidase (MAO), which is responsible for the metabolism of monoamines. MC treatment reduces levels of plasma corticosterone and restores hippocampal BDNF (full form) protein in stressed animals. Further, MC has shown its protective effect against amyloid-beta, MPTP, rotenone, 6-OHDA, etc. suggesting its potential role against neurodegenerative disorders. The aim of the present review is to highlight the therapeutic potential of MC in the treatment of several neurological, neuropsychiatric, and neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Neha Ghiloria
- Dr. Baba Saheb Ambedkar Hospital, Rohini, New Delhi 110085, India
| | | | - Dheeraj Bisht
- Department of Pharmaceutical Sciences, Sir J.C. Bose Technical Campus, Bhimtal, Kumaun University, Nainital, Uttarakhand 263002, India
| | | | - Vaibhav Walia
- Department of Pharmacology, SGT College of Pharmacy, SGT University, Gurugram, Haryana 122505, India
| | - Md Sabir Alam
- Department of Pharmaceutics, SGT College of Pharmacy, SGT University, Gurugram, Haryana 122505, India
| |
Collapse
|
12
|
Cruz ASD, Drehmer MM, Baetas-da-Cruz W, Machado JC. Ultrasound biomicroscopy in the quantification of brain perfusion parameters of a rat stroke model: Analysis of contrast agent bolus kinetic dynamics. J Neurosci Methods 2024; 401:110005. [PMID: 37931754 DOI: 10.1016/j.jneumeth.2023.110005] [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: 07/28/2023] [Revised: 10/23/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Ischemic stroke represents a significant global health concern, necessitating thorough investigations and the utilization of stroke animal models to explore novel treatment modalities and diagnostic imaging techniques. NEW METHOD Ultrasound biomicroscopy (BMU), operating at a center frequency of 21 MHz, along with ultrasound contrast agents (UCAs), was used to quantify microcirculation cerebral blood flow in a rat model of ischemic stroke. The microcirculation parameters were derived from time intensity curve (TIC) plots obtained based on UCA-bolus kinetics. RESULTS Semiquantitative perfusion-related parameters were assessed. The TIC curves showed differences in amplitude when compared intra-animal between the left and right sides, and three situations were observed: normal perfusion, hypoperfusion, and nonperfusion. ROC analysis of delays between the left and right time intensity peak (TIP) for regions of interest (ROIs) in the control and stroke-hypoperfusion groups revealed an optimal cutpoint of 0.39 s to indicate when hypoperfusion is occurring in rats, with a sensitivity of 93.33 % and a specificity of 80 %. COMPARISON WITH EXISTING METHOD(S) Ultrasound perfusion imaging through the temporal bone window has been clinically applied to stroke patients using a UCA bolus for TIC analysis. TIC parameters were correlated with MRI- and CT-based measurements. CONCLUSIONS This investigation quantified cerebral blood flow in a rat model of ischemic stroke by measuring microcirculation parameters. The study demonstrated the efficacy of this approach as a valuable tool for conducting preclinical studies.
Collapse
Affiliation(s)
- Aline Silva da Cruz
- Biomedical Engineering Program, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Maria Margarida Drehmer
- Post-Graduation Program in Surgical Sciences, Department of Surgery, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Wagner Baetas-da-Cruz
- Post-Graduation Program in Surgical Sciences, Department of Surgery, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - João Carlos Machado
- Biomedical Engineering Program, COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Post-Graduation Program in Surgical Sciences, Department of Surgery, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
13
|
Simon Machado R, Mathias K, Joaquim L, Willig de Quadros R, Petronilho F, Tezza Rezin G. From diabetic hyperglycemia to cerebrovascular Damage: A narrative review. Brain Res 2023; 1821:148611. [PMID: 37793604 DOI: 10.1016/j.brainres.2023.148611] [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: 07/13/2023] [Revised: 09/04/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023]
Abstract
Diabetes mellitus is a globally significant disease that can lead to systemic complications, particularly vascular damage, including cardiovascular and cerebrovascular diseases of relevance. The physiological changes resulting from the imbalance in blood glucose levels play a crucial role in initiating vascular endothelial damage. Elevated glucose levels can also penetrate the central nervous system, triggering diabetic encephalopathy characterized by oxidative damage to brain components and activation of alternative and neurotoxic pathways. This brain damage increases the risk of ischemic stroke, a leading cause of mortality worldwide and a major cause of disability among surviving patients. The aim of this review is to highlight important pathways related to hyperglycemic damage that extend to the brain and result in vascular dysfunction, ultimately leading to the occurrence of a stroke. Understanding how diabetes mellitus contributes to the development of ischemic stroke and its impact on patient outcomes is crucial for implementing therapeutic strategies that reduce the incidence of diabetes mellitus and its complications, ultimately decreasing morbidity and mortality associated with the disease.
Collapse
Affiliation(s)
- Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil; Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil.
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Larissa Joaquim
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Rafaella Willig de Quadros
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| |
Collapse
|
14
|
Lu G, Su X, Wang L, Leung CK, Zhou J, Xiong Z, Wang W, Liu H, Chan WY. Neuroprotective Effects of Human-Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cell Extracellular Vesicles in Ischemic Stroke Models. Biomedicines 2023; 11:2550. [PMID: 37760991 PMCID: PMC10525838 DOI: 10.3390/biomedicines11092550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Stroke represents the second leading cause of death and the primary cause of long-term disability in humans. The transplantation of mesenchymal stem cells (MSC) reportedly improves functional outcomes in animal models of cerebral ischemia. Here, we evaluate the neuroprotective potential of extracellular vesicles secreted from human-induced pluripotent stem cell-derived mesenchymal stem cells (hiPS-MSC-EV) using preclinical cell-based and animal-based models of ischemic strokes. METHODS hiPS-MSC-EV were isolated using an ultrafiltration method. HT22 cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) injury for 2 h, followed by treatment with hiPS-MSC-EV (100 μg/mL). Male C57BL/6 mice were subjected to middle cerebral artery occlusion (MCAO) followed by an intravenous injection of hiPS-MSC-EV (100 μg) at three distinct time points. RESULTS Our experimental approach revealed hiPS-MSC-EV promoted HT22 cell proliferation, reduced apoptosis, and altered cellular morphology following OGD/R. In addition, hiPS-MSC-EV reduced the volume of infarcts, improved spontaneous movement abilities, and enhanced angiogenesis by expressing the VEGF and CXCR4 proteins in the infarcted hemisphere of the MCAO-treated mouse model. CONCLUSION Our findings provide evidence of the potential neuroprotective effects of hiPS-MSC-derived extracellular vesicles (hiPS-MSC-EVs) in both in vitro and in vivo mouse models of ischemic stroke. These results suggest that hiPS-MSC-EVs may play a role in neurorestoration and offer insights into potential cell-free strategies for addressing cerebral ischemia.
Collapse
Affiliation(s)
- Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xianwei Su
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
| | - Lihong Wang
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
| | - Chi-Kwan Leung
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
| | - Jingye Zhou
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
| | - Zhiqiang Xiong
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China; (Z.X.); (H.L.)
| | - Wuming Wang
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan 250012, China; (Z.X.); (H.L.)
| | - Wai-Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; (G.L.); (X.S.); (L.W.); (J.Z.); (W.W.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
15
|
Choi YH, Hsu M, Laaker C, Herbath M, Yang H, Cismaru P, Johnson AM, Spellman B, Wigand K, Sandor M, Fabry Z. Dual role of Vascular Endothelial Growth Factor-C (VEGF-C) in post-stroke recovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555144. [PMID: 37693558 PMCID: PMC10491156 DOI: 10.1101/2023.08.30.555144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Using a mouse model of ischemic stroke, this study characterizes stroke-induced lymphangiogenesis at the cribriform plate (CP). While blocking CP lymphangiogenesis with a VEGFR-3 inhibitor improves stroke outcome, administration of VEGF-C induced larger brain infarcts. Abstract Cerebrospinal fluid (CSF), antigens, and antigen-presenting cells drain from the central nervous system (CNS) into lymphatic vessels near the cribriform plate and dural meningeal lymphatics. However, the pathological roles of these lymphatic vessels surrounding the CNS during stroke are not well understood. Using a mouse model of ischemic stroke, transient middle cerebral artery occlusion (tMCAO), we show that stroke induces lymphangiogenesis near the cribriform plate. Interestingly, lymphangiogenesis is restricted to lymphatic vessels at the cribriform plate and downstream cervical lymph nodes, without affecting the conserved network of lymphatic vessels in the dura. Cribriform plate lymphangiogenesis peaks at day 7 and regresses by day 14 following tMCAO and is regulated by VEGF-C/VEGFR-3. These newly developed lymphangiogenic vessels transport CSF and immune cells to the cervical lymph nodes. Inhibition of VEGF-C/VEGFR-3 signaling using a blocker of VEGFR-3 prevented lymphangiogenesis and led to improved stroke outcomes at earlier time points but had no effects at later time points following stroke. Administration of VEGF-C after tMCAO did not further increase post-stroke lymphangiogenesis, but instead induced larger brain infarcts. The differential roles for VEGFR-3 inhibition and VEGF-C in regulating stroke pathology call into question recent suggestions to use VEGF-C therapeutically for stroke.
Collapse
|
16
|
Xu Q, Cheung RTF. Melatonin mitigates type 1 diabetes-aggravated cerebral ischemia-reperfusion injury through anti-inflammatory and anti-apoptotic effects. Brain Behav 2023; 13:e3118. [PMID: 37327371 PMCID: PMC10498092 DOI: 10.1002/brb3.3118] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/15/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
INTRODUCTION Cerebral ischemia and diabetes mellitus (DM) are common diseases that often coexist and interact with each other. DM doubles the risk of ischemic stroke, and cerebral ischemia causes stress-induced hyperglycemia. Most experimental stroke studies used healthy animals. Melatonin is neuroprotective against cerebral ischemia-reperfusion injury (CIRI) in non-DM, normoglycemic animals through anti-oxidant effect, anti-inflammation, and anti-apoptosis. Previous studies have also reported a negative correlation between hyperglycemia and urinary melatonin metabolite. OBJECTIVES The present study investigated the effects of type 1 DM (T1DM) on CIRI in rats and the role of melatonin against CIRI in T1DM animals. RESULTS Our results revealed that T1DM aggravated CIRI, leading to greater weight loss, increased infarct volume, and worse neurological deficit. T1DM aggravated the post-CIRI activation of nuclear factor kappa B (NF-κB) pathway and increase in pro-apoptotic markers. A single intraperitoneal injection of melatonin at 10 mg/kg given 30 min before ischemia onset attenuated CIRI in T1DM rats, resulting in less weight loss, decreased infarct volume, and milder neurological deficit when compared with the vehicle group. Melatonin treatment achieved anti-inflammatory and anti-apoptotic effects with reduced NF-κB pathway activation, reduced mitochondrial cytochrome C release, decreased calpain-mediated spectrin breakdown product (SBDP), and decreased caspase-3-mediated SBDP. The treatment also led to fewer iNOS+ cells, milder CD-68+ macrophage/microglia infiltration, decreased TUNEL+ apoptotic cells, and better neuronal survival. CONCLUSIONS T1DM aggravates CIRI. Melatonin treatment is neuroprotective against CIRI in T1DM rats via anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Qian Xu
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Raymond Tak Fai Cheung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| |
Collapse
|
17
|
Choi SG, Shin J, Lee KY, Park H, Kim SI, Yi YY, Kim DW, Song HJ, Shin HJ. PINK1 siRNA-loaded poly(lactic-co-glycolic acid) nanoparticles provide neuroprotection in a mouse model of photothrombosis-induced ischemic stroke. Glia 2023; 71:1294-1310. [PMID: 36655313 DOI: 10.1002/glia.24339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023]
Abstract
PTEN-induced kinase 1 (PINK1) is a well-known critical marker in the pathway for mitophagy regulation as well as mitochondrial dysfunction. Evidence suggests that mitochondrial dynamics and mitophagy flux play an important role in the development of brain damage from stroke pathogenesis. In this study, we propose a treatment strategy using nanoparticles that can control PINK1. We used a murine photothrombotic ischemic stroke (PTS) model in which clogging of blood vessels is induced with Rose Bengal (RB) to cause brain damage. We targeted PINK1 with poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles loaded with PINK1 siRNA (PINK1 NPs). After characterizing siRNA loading in the nanoparticles, we assessed the efficacy of PINK1 NPs in mice with PTS using immunohistochemistry, 1% 2,3,5-triphenyltetrazolium chloride staining, measurement of motor dysfunction, and Western blot. PINK1 was highly expressed in microglia 24 h after PTS induction. PINK1 siRNA treatment increased phagocytic activity, migration, and expression of an anti-inflammatory state in microglia. In addition, the PLGA nanoparticles were selectively taken up by microglia and specifically regulated PINK1 expression in those cells. Treatment with PINK1 NPs prior to stroke induction reduced expression of mitophagy-inducing factors, infarct volume, and motor dysfunction in mice with photothrombotic ischemia. Experiments with PINK1-knockout mice and microglia depletion with PLX3397 confirmed a decrease in stroke-induced infarct volume and behavioral dysfunction. Application of nanoparticles for PINK1 inhibition attenuates RB-induced photothrombotic ischemic injury by inhibiting microglia responses, suggesting that a nanomedical approach targeting the PINK1 pathway may provide a therapeutic avenue for stroke treatment.
Collapse
Affiliation(s)
- Seung Gyu Choi
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ka Young Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Yoon Young Yi
- Department of Pediatrics, College of Medicine, Hallym University and Gangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea
- Department of Anatomy and Cell Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University Sejong Hospital and College of Medicine, Republic of Korea
| | - Hyo Jung Shin
- Brain Research Institute, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
18
|
Zeng F, Cao J, Hong Z, Liu Y, Hao J, Qin Z, Zou X, Tao T. Single-cell analyses reveal the dynamic functions of Itgb2 + microglia subclusters at different stages of cerebral ischemia-reperfusion injury in transient middle cerebral occlusion mice model. Front Immunol 2023; 14:1114663. [PMID: 37063847 PMCID: PMC10098327 DOI: 10.3389/fimmu.2023.1114663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/28/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction The underlying pathophysiological mechanisms of cerebral ischemia reperfusion injury (CIRI) is intricate, and current studies suggest that neuron, astrocyte, microglia, endothelial cell, and pericyte all have different phenotypic changes of specific cell types after ischemic stroke. And microglia account for the largest proportion after CIRI. Previous transcriptomic studies of ischemic stroke have typically focused on the 24 hours after CIRI, obscuring the dynamics of cellular subclusters throughout the disease process. Therefore, traditional methods for identifying cell types and their subclusters may not be sufficient to fully unveil the complexity of single-cell transcriptional profile dynamics caused by an ischemic stroke. Methods In this study, to explore the dynamic transcriptional profile of single cells after CIRI, we used single-cell State Transition Across-samples of RNA-seq data (scSTAR), a new bioinformatics method, to analyze the single-cell transcriptional profile of day 1, 3, and 7 of transient middle cerebral artery occlusion (tMCAO) mice. Combining our bulk RNA sequences and proteomics data, we found the importance of the integrin beta 2 (Itgb2) gene in post-modeling. And microglia of Itgb2+ and Itgb2- were clustered by the scSTAR method. Finally, the functions of the subpopulations were defined by Matescape, and three different time points after tMCAO were found to exhibit specific functions. Results Our analysis revealed a dynamic transcriptional profile of single cells in microglia after tMCAO and explored the important role of Itgb2 contributed to microglia by combined transcriptomics and proteomics analysis after modeling. Our further analysis revealed that the Itgb2+ microglia subcluster was mainly involved in energy metabolism, cell cycle, angiogenesis, neuronal myelin formation, and repair at 1, 3, and 7 days after tMCAO, respectively. Discussion Our results suggested that Itgb2+ microglia act as a time-specific multifunctional immunomodulatory subcluster during CIRI, and the underlying mechanisms remain to be further investigated.
Collapse
Affiliation(s)
- Fanning Zeng
- Department of Anesthesiology, Central People’s Hospital of Zhanjiang, Zhanjiang, Guangdong, China
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Cao
- Department of Anesthesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zexuan Hong
- Department of Anesthesiology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yujun Liu
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Hao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zaisheng Qin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Zou
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Tao Tao
- Department of Anesthesiology, Central People’s Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| |
Collapse
|
19
|
Influence of sex, age and diabetes on brain transcriptome and proteome modifications following cerebral ischemia. BMC Neurosci 2023; 24:7. [PMID: 36707762 PMCID: PMC9881265 DOI: 10.1186/s12868-023-00775-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Ischemic stroke is a major cause of death and disability worldwide. Translation into the clinical setting of neuroprotective agents showing promising results in pre-clinical studies has systematically failed. One possible explanation is that the animal models used to test neuroprotectants do not properly represent the population affected by stroke, as most of the pre-clinical studies are performed in healthy young male mice. Therefore, we aimed to determine if the response to cerebral ischemia differed depending on age, sex and the presence of comorbidities. Thus, we explored proteomic and transcriptomic changes triggered during the hyperacute phase of cerebral ischemia (by transient intraluminal middle cerebral artery occlusion) in the brain of: (1) young male mice, (2) young female mice, (3) aged male mice and (4) diabetic young male mice. Moreover, we compared each group's proteomic and transcriptomic changes using an integrative enrichment pathways analysis to disclose key common and exclusive altered proteins, genes and pathways in the first stages of the disease. We found 61 differentially expressed genes (DEG) in male mice, 77 in females, 699 in diabetics and 24 in aged mice. Of these, only 14 were commonly dysregulated in all groups. The enrichment pathways analysis revealed that the inflammatory response was the biological process with more DEG in all groups, followed by hemopoiesis. Our findings indicate that the response to cerebral ischemia regarding proteomic and transcriptomic changes differs depending on sex, age and comorbidities, highlighting the importance of incorporating animals with different phenotypes in future stroke research studies.
Collapse
|
20
|
Wong A, Bhuiyan MIH, Rothman J, Drew K, Pourrezaei K, Sun D, Barati Z. Near infrared spectroscopy detection of hemispheric cerebral ischemia following middle cerebral artery occlusion in rats. Neurochem Int 2023; 162:105460. [PMID: 36455748 PMCID: PMC10263189 DOI: 10.1016/j.neuint.2022.105460] [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: 08/31/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Timely and sensitive in vivo estimation of ischemic stroke-induced brain infarction are necessary to guide diagnosis and evaluation of treatments' efficacy. The gold standard for estimation of the cerebral infarction volume is magnetic resonance imaging (MRI), which is expensive and not readily accessible. Measuring regional cerebral blood flow (rCBF) with Laser Doppler flowmetry (LDF) is the status quo for confirming reduced blood flow in experimental ischemic stroke models. However, rCBF reduction following cerebral artery occlusion often does not correlate with subsequent infarct volume. In the present study, we employed the continuous-wave near infrared spectroscopy (NIRS) technique to monitor cerebral oxygenation during 90 min of the intraluminal middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats (n = 8, male). The NIRS device consisted of a controller module and an optical sensor with two LED light sources and two photodiodes making up two parallel channels for monitoring left and right cerebral hemispheres. Optical intensity measurements were converted to deoxyhemoglobin (Hb) and oxyhemoglobin (HbO2) changes relative to a 2-min window prior to MCAO. Area under the curve (auc) for Hb and HbO2 was calculated for the 90-min occlusion period for each hemisphere (ipsilateral and contralateral). To obtain a measure of total ischemia, auc of the contralateral side was subtracted from the ipsilateral side resulting in ΔHb and ΔHbO2 parameters. Infarct volume (IV) was calculated by triphenyl tetrazolium chloride (TTC) staining at 24h reperfusion. Results showed a significant negative correlation (r = -0.81, p = 0.03) between ΔHb and infarct volume. In conclusion, our results show feasibility of using a noninvasive optical imaging instrument, namely NIRS, in monitoring cerebral ischemia in a rodent stroke model. This cost-effective, non-invasive technique may improve the rigor of experimental models of ischemic stroke by enabling in vivo longitudinal assessment of cerebral oxygenation and ischemic injury.
Collapse
Affiliation(s)
- Ardy Wong
- Drexel University School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA, USA
| | - Mohammad Iqbal Hossain Bhuiyan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, PA, 15260, USA; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pennsylvania, PA, 15260, USA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX, 79968, USA
| | | | - Kelly Drew
- Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, USA
| | - Kambiz Pourrezaei
- Drexel University School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, PA, 15260, USA; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pennsylvania, PA, 15260, USA
| | - Zeinab Barati
- Barati Medical LLC, Fairbanks, AK, USA; Center for Transformative Research in Metabolism, Institute of Arctic Biology, University of Alaska Fairbanks, USA.
| |
Collapse
|
21
|
Amado B, Melo L, Pinto R, Lobo A, Barros P, Gomes JR. Ischemic Stroke, Lessons from the Past towards Effective Preclinical Models. Biomedicines 2022; 10:2561. [PMID: 36289822 PMCID: PMC9599148 DOI: 10.3390/biomedicines10102561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
Ischemic stroke is a leading cause of death worldwide, mainly in western countries. So far, approved therapies rely on reperfusion of the affected brain area, by intravenous thrombolysis or mechanical thrombectomy. The last approach constitutes a breakthrough in the field, by extending the therapeutic window to 16-24 h after stroke onset and reducing stroke mortality. The combination of pharmacological brain-protective strategies with reperfusion is the future of stroke therapy, aiming to reduce brain cell death and decrease patients' disabilities. Recently, a brain-protective drug-nerinetide-reduced brain infarct and stroke mortality, and improved patients' functional outcomes in clinical trials. The success of new therapies relies on bringing preclinical studies and clinical practice close together, by including a functional outcome assessment similar to clinical reality. In this review, we focused on recent upgrades of in vitro and in vivo stroke models for more accurate and effective evaluation of therapeutic strategies: from spheroids to organoids, in vitro models that include all brain cell types and allow high throughput drug screening, to advancements in in vivo preclinical mouse stroke models to mimic the clinical reality in surgical procedures, postsurgical care, and functional assessment.
Collapse
Affiliation(s)
- Beatriz Amado
- Molecular Neurobiology Group, IBMC—Instituto de Biologia Molecular e Celular, 4200-135 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Lúcia Melo
- Molecular Neurobiology Group, IBMC—Instituto de Biologia Molecular e Celular, 4200-135 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Raquel Pinto
- Molecular Neurobiology Group, IBMC—Instituto de Biologia Molecular e Celular, 4200-135 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | | | - Pedro Barros
- Neurology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, 4434-502 Vila Nova de Gaia, Portugal
- Stroke Unit, Centro Hospitalar de Vila Nova de Gaia/Espinho, 4434-502 Vila Nova de Gaia, Portugal
| | - João R. Gomes
- Molecular Neurobiology Group, IBMC—Instituto de Biologia Molecular e Celular, 4200-135 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| |
Collapse
|
22
|
Friedrich J, Lindauer U, Höllig A. Procedural and Methodological Quality in Preclinical Stroke Research-A Cohort Analysis of the Rat MCAO Model Comparing Periods Before and After the Publication of STAIR/ARRIVE. Front Neurol 2022; 13:834003. [PMID: 35707032 PMCID: PMC9190283 DOI: 10.3389/fneur.2022.834003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 04/12/2022] [Indexed: 11/24/2022] Open
Abstract
The translation of preclinical stroke research into successful human clinical trials remains a challenging task. The first Stroke Therapy Academic Industry Roundtable (STAIR) recommendations for preclinical research and several other guidelines were published to address these challenges. Most guidelines recommend the use of physiological monitoring to detect the occurrence of undesired pathologies such as subarachnoid hemorrhage and to limit the variability of the infarct volume and–therefore-homogenize the experimental result for complete reporting particularly with respect to transparency and methodological rigor. From the years 2009 and 2019, 100 published articles each using a rat stroke model were analyzed to quantify parameters related to anesthesia, physiological monitoring, stroke model type, ischemia verification, and overall study quality over time. No significant difference in the frequency of cerebral blood flow (CBF) measurements over time (28/34% for 2009/2019) was found. Notably, significantly fewer studies reported temperature, blood pressure, and blood gas monitoring data in 2019 compared to 2009. On the other hand, an increase in general study quality parameters (e.g., randomization, reporting of approval) was seen. In conclusion, the frequency of periinterventional monitoring has decreased over time. Some general methodological quality aspects, however, partially have increased. CBF measurement–the gold standard for ischemia verification-was applied rarely. Despite the growing recognition of current guidelines such as STAIR and ARRIVE (both widely approved in 2019) reporting, methods and procedures mostly do not follow these guidelines. These deficits may contribute to the translational failure of preclinical stroke research in search for neuroprotective therapies.
Collapse
Affiliation(s)
| | - Ute Lindauer
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
23
|
Xin YY, Wang JX, Xu AJ. Electroacupuncture ameliorates neuroinflammation in animal models. Acupunct Med 2022; 40:474-483. [PMID: 35229660 DOI: 10.1177/09645284221076515] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Neuroinflammation refers to a wide range of immune responses occurring in the brain or spinal cord. It is closely related to a variety of neurodegenerative diseases, for which it potentially represents a new direction for treatment. Electroacupuncture (EA) is one method of acupuncture treatment, which can be used as an adjuvant therapy for many diseases. This review focuses on molecular mechanisms of EA in the reduction of neuroinflammation, summarizes relevant basic research and outlines future directions for investigation. Findings: A growing body of basic research has shown that EA can ameliorate neuroinflammation centrally (in animal models of ischemic stroke, Alzheimer’s disease, traumatic brain injury, spinal cord injury, Parkinson’s disease and vascular dementia) and peripherally (e.g. after a surgical insult or injection of lipopolysaccharide) and that its effects involve different molecular mechanisms, including activation of the α7 nicotinic acetylcholine receptor signaling pathway and P2 type purinergic receptors, inhibition of nuclear factor κB, and mitigation of damage secondary to oxidative stress and NOD-like receptor protein 3 inflammasome activation. Conclusions: EA is capable of regulating multiple cell signal transduction pathways to alleviate neuroinflammation in animal models. Although the findings of animal studies are encouraging, further prospective clinical trials are needed to verify the efficacy of EA for the treatment of neuroinflammation.
Collapse
Affiliation(s)
- Yue-yang Xin
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-xu Wang
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ai-jun Xu
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Lessons Learned from Phase II and Phase III Trials Investigating Therapeutic Agents for Cerebral Ischemia Associated with Aneurysmal Subarachnoid Hemorrhage. Neurocrit Care 2021; 36:662-681. [PMID: 34940927 DOI: 10.1007/s12028-021-01372-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 10/04/2021] [Indexed: 12/20/2022]
Abstract
One of the challenges in bringing new therapeutic agents (since nimodipine) in for the treatment of cerebral ischemia associated with aneurysmal subarachnoid hemorrhage (aSAH) is the incongruence in therapeutic benefit observed between phase II and subsequent phase III clinical trials. Therefore, identifying areas for improvement in the methodology and interpretation of results is necessary to increase the value of phase II trials. We performed a systematic review of phase II trials that continued into phase III trials, evaluating a therapeutic agent for the treatment of cerebral ischemia associated with aSAH. We followed the Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines for systematic reviews, and review was based on a peer-reviewed protocol (International Prospective Register of Systematic Reviews no. 222965). A total of nine phase III trials involving 7,088 patients were performed based on eight phase II trials involving 1558 patients. The following therapeutic agents were evaluated in the selected phase II and phase III trials: intravenous tirilazad, intravenous nicardipine, intravenous clazosentan, intravenous magnesium, oral statins, and intraventricular nimodipine. Shortcomings in several design elements of the phase II aSAH trials were identified that may explain the incongruence between phase II and phase III trial results. We suggest the consideration of the following strategies to improve phase II design: increased focus on the selection of surrogate markers of efficacy, selection of the optimal dose and timing of intervention, adjustment for exaggerated estimate of treatment effect in sample size calculations, use of prespecified go/no-go criteria using futility design, use of multicenter design, enrichment of the study population, use of concurrent control or placebo group, and use of innovative trial designs such as seamless phase II to III design. Modifying the design of phase II trials on the basis of lessons learned from previous phase II and phase III trial combinations is necessary to plan more effective phase III trials.
Collapse
|
25
|
Mao Y, Qu Y, Wang Q. Cryptotanshinone reduces neurotoxicity induced by cerebral ischemia-reperfusion injury involving modulation of microglial polarization. Restor Neurol Neurosci 2021; 39:209-220. [PMID: 34219678 DOI: 10.3233/rnn-201070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND The diterpenoid cryptotanshinone (CTS) has wide biological functions, including inhibition of tumor growth, inflammation and apoptosis. The present study aimed to explore the possible effect of CTS on cerebral ischemia/reperfusion (I/R) injury and the underlying mechanisms. METHODS Male C57BL/6J mice underwent transient middle cerebral artery occlusion (tMCAO) and murine microglia BV2 cells were challenged by Oxygen/glucose deprivation, to mimic I/R and ischemic/hypoxic and reperfusion (H/R) injury, respectively. CTS was administered 0.5 h (10 mg/kg) after the onset of MCAO or 2 h (20μM) post OGD. Infarct volume and neurological deficit were measured. Immunofluorescence, qPCR, and western blot, were performed to detect the expression of cytokines, apoptotic marker, and M1/M2 phenotype-specific genes. Flow cytometry was applied for M1/M2 subpopulation or Annexin V/PI apoptosis assessment. RESULTS CTS significantly reduced cerebral infarct volume, neurologic deficit scores, pro-inflammatory cytokine production (IL-6, TNF-α, and IL-1β), apoptotic protein expression (cleaved caspase-3) of mice after tMCAO challenge. Furthermore, CTS attenuated CD16+ M1-type and elevated CD206+ M2-type microglia in vivo or in vitro. CONCLUSIONS We propose that the neuroprotective effect of CTS in the I/R or H/R context are explained modulation of microglial polarization, suggesting therapeutic potential for cerebral ischemic stroke.
Collapse
Affiliation(s)
- Yanfang Mao
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Yang Qu
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Qingdong Wang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong, China
| |
Collapse
|
26
|
Abstract
Neurodegenerative diseases, characterized by progressive neural loss, have been some of the most challenging medical problems in aging societies. Treatment strategies such as symptom management have little impact on dis-ease progression, while intervention with specific disease mechanisms may only slow down disease progression. One therapeutic strategy that has the potential to reverse the disease phenotype is to replenish neurons and re-build the pathway lost to degeneration. Although it is generally believed that the central nervous system has lost the capability to regenerate, increasing evidence indicates that the brain is more plastic than previously thought, containing perhaps the biggest repertoire of cells with latent neurogenic programs in the body. This review focuses on key advances in generating new neurons through in situ neuronal reprogramming, which is tied to fun-damental questions regarding adult neurogenesis, cell source, and mecha-nisms for neuronal reprogramming, as well as the ability of new neurons to integrate into the existing circuitry. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Hao Qian
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0651, USA;
| | - Xiang-Dong Fu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0651, USA;
| |
Collapse
|
27
|
Komatsu T, Ohta H, Motegi H, Hata J, Terawaki K, Koizumi M, Muta K, Okano HJ, Iguchi Y. A novel model of ischemia in rats with middle cerebral artery occlusion using a microcatheter and zirconia ball under fluoroscopy. Sci Rep 2021; 11:12806. [PMID: 34140618 PMCID: PMC8211726 DOI: 10.1038/s41598-021-92321-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/04/2021] [Indexed: 12/21/2022] Open
Abstract
The failure of neuroprotective treatment-related clinical trials may be partially caused by unestablished animal models. Existing animal models are less likely to provide occlusion confined to the middle cerebral artery (MCA), making transarterial intervention difficult. We aimed to develop a novel focal stroke model using a microcatheter and zirconium dioxide that is non-magnetic under fluoroscopic guidance, which can monitor MCA occlusion and can improve hemorrhagic complications. Using male Sprague Dawley rats (n = 10), a microcatheter was navigated from the caudal ventral artery to the left internal carotid artery using an X-ray fluoroscopy to establish local occlusion. All rat cerebral angiographies were successful. No rats had hemorrhagic complications. Eight (80%) rats underwent occlusion of the MCA bifurcation by zirconium dioxide. Accidentally, the left posterior cerebral artery was failure embolized in 2 rats (20%). The median operating time was 8 min. All rats of occlusion MCA revealed an incomplete hemiparesis on the right side with neurological deficit score ranging from 1 to 3 (median 1, interquartile range 1-3) at 24 h after the induction of ischemia. Moreover, 2% 2,3,5-triphenyl tetrazolium chloride staining showed that the median infarct volume (mm3) was 280 (interquartile range 267-333) 24 h after the left MCA bifurcation occlusion. We present a novel rat model for focal stroke using a microcatheter and zirconium dioxide which does not affect the MRI. The model is predictable which is well confined within the territory supplied by the MCA, and reproducibility of this model is 80%. Fluoroscopy was able to identify which the MCA occlusion and model success while creating the model. It permitted exclusion of animals with complications from the experiment.
Collapse
Affiliation(s)
- Teppei Komatsu
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Hiroki Ohta
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Haruhiko Motegi
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Junichi Hata
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Koshiro Terawaki
- Department of Radiological Science, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Makoto Koizumi
- Laboratory Animal Facilities, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Kanako Muta
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, 3-25-8 Nishishimbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
28
|
Novel brain-targeting 3-n-butylphthalide prodrugs for ischemic stroke treatment. J Control Release 2021; 335:498-514. [PMID: 34087248 DOI: 10.1016/j.jconrel.2021.05.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Currently, ischemic stroke is the leading cause of disability and death worldwide, and the performance of corresponding drugs is often unsatisfactory owing to the complex pathological processes and the impediment of the blood-brain barrier (BBB). Here, we employed various tertiary amino groups, including different linear, cyclic, and bimolecular drug structures, to modify 3-n-butylphthalide (NBP), a natural product used for ischemic stroke treatment, which has poor bioavailability, to generate a series of six prodrugs. These prodrugs showed significantly improved solubility and cellular uptake, which were primarily driven by putative pyrilamine cationic transporters. They also displayed more efficient brain delivery in vivo, reaching as high as 21.5-fold brain accumulation increase compared with NBP, leading to much higher bioavailability and stronger therapeutic effects. The toxicity of these molecules is also lower or similar to that of unmodified NBP. We showed that the tertiary amino group-modified NBP prodrugs are effective and safe for treating ischemic stroke with significantly enhanced druggability; hence, they have potential for further clinical development.
Collapse
|
29
|
Hong SH, Hong JH, Lahey MT, Zhu L, Stephenson JM, Marrelli SP. A low-cost mouse cage warming system provides improved intra-ischemic and post-ischemic body temperature control - Application for reducing variability in experimental stroke studies. J Neurosci Methods 2021; 360:109228. [PMID: 34052289 DOI: 10.1016/j.jneumeth.2021.109228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Brain temperature is a strong determinant of ischemic stroke injury. For this reason, tight management of brain or body temperature (Tcore) in experimental rodent stroke models is recommended to improve the rigor and reproducibility of outcomes. However, methods for managing Tcore during and after stroke vary widely in approach and effectiveness. NEW METHOD We developed a low-cost warm ambient air cage (WAAC) system to provide improved temperature control during the intra-ischemic and post-ischemic recovery periods. The system is incorporated into standard holding cages for maintaining Tcore during the intra-ischemic period as well as for several hours into the recovery period. RESULTS AND COMPARISON WITH EXISTING METHODS We compared the WAAC system with a commonly used heat support method, consisting of a cage on a heating pad. Both heat support systems were evaluated for the middle cerebral artery occlusion (MCAo) stroke model in mice. The WAAC system provided improved temperature control (more normothermic Tcore and less Tcore variation) during the intra- ischemic period (60 min) and post-ischemic period (3 h). Mean infarct volume was not statistically different by heat support system, however, standard deviation was 54 % lower in the WAAC system group. CONCLUSIONS Mice and other small rodents are highly vulnerable to heat loss during and after the MCAo procedure. The WAAC system provides more precise and controlled Tcore maintenance compared with frequently used induction heating methods in mice undergoing the MCAo stroke model. The improved temperature control should enhance experimental rigor and reduce the number of experimental animals needed.
Collapse
Affiliation(s)
- Sung-Ha Hong
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Jeong-Ho Hong
- Department of Neurology, Brain Research Institute, Keimyung University School of Medicine, Dongsan Medical Center, Daegu, South Korea
| | - Matthew T Lahey
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Liang Zhu
- Department of Medicine, McGovern Medical School, the University of Texas Health Science Center, Houston, TX, USA
| | - Jessica M Stephenson
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Sean P Marrelli
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
30
|
Singh D, Wasan H, Reeta KH. Preclinical Stroke Research and Translational Failure: A Bird's Eye View on Preventable Variables. Cell Mol Neurobiol 2021; 42:2003-2017. [PMID: 33786698 DOI: 10.1007/s10571-021-01083-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/18/2021] [Indexed: 02/08/2023]
Abstract
Despite achieving remarkable success in understanding the cellular, molecular and pathophysiological aspects of stroke, translation from preclinical research has always remained an area of debate. Although thousands of experimental compounds have been reported to be neuro-protective, their failures in clinical setting have left the researchers and stakeholders in doldrums. Though the failures described have been excruciating, they also give us a chance to refocus on the shortcomings. For better translational value, evidences from preclinical studies should be robust and reliable. Preclinical study design has a plethora of variables affecting the study outcome. Hence, this review focusses on the factors to be considered for a well-planned preclinical study while adhering to guidelines with emphasis on the study design, commonly used animal models, their limitations with special attention on various preventable attritions including comorbidities, aged animals, time of dosing, outcome measures and physiological variables along with the concept of multicentric preclinical randomized controlled trials. Here, we provide an overview of a panorama of practical aspects, which could be implemented, so that a well-defined preclinical study would result in a neuro-protectant with better translational value.
Collapse
Affiliation(s)
- Devendra Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Himika Wasan
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
31
|
Trotman-Lucas M, Gibson CL. A review of experimental models of focal cerebral ischemia focusing on the middle cerebral artery occlusion model. F1000Res 2021; 10:242. [PMID: 34046164 PMCID: PMC8127011 DOI: 10.12688/f1000research.51752.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemic stroke is a leading cause of death and disability, but current pharmacological therapies are limited in their utility and effectiveness.
In vitro and
in vivo models of ischemic stroke have been developed which allow us to further elucidate the pathophysiological mechanisms of injury and investigate potential drug targets.
In vitro models permit mechanistic investigation of the biochemical and molecular mechanisms of injury but are reductionist and do not mimic the complexity of clinical stroke.
In vivo models of ischemic stroke directly replicate the reduction in blood flow and the resulting impact on nervous tissue. The most frequently used
in vivo model of ischemic stroke is the intraluminal suture middle cerebral artery occlusion (iMCAO) model, which has been fundamental in revealing various aspects of stroke pathology. However, the iMCAO model produces lesion volumes with large standard deviations even though rigid surgical and data collection protocols are followed. There is a need to refine the MCAO model to reduce variability in the standard outcome measure of lesion volume. The typical approach to produce vessel occlusion is to induce an obstruction at the origin of the middle cerebral artery and reperfusion is reliant on the Circle of Willis (CoW). However, in rodents the CoW is anatomically highly variable which could account for variations in lesion volume. Thus, we developed a refined approach whereby reliance on the CoW for reperfusion was removed. This approach improved reperfusion to the ischemic hemisphere, reduced variability in lesion volume by 30%, and reduced group sizes required to determine an effective treatment response by almost 40%. This refinement involves a methodological adaptation of the original surgical approach which we have shared with the scientific community via publication of a visualised methods article and providing hands-on training to other experimental stroke researchers.
Collapse
Affiliation(s)
| | - Claire L Gibson
- School of Psychology, University of Nottingham, Nottingham, NG7 2UH, UK
| |
Collapse
|
32
|
Guo Y, Zhou J, Li X, Xiao Y, Zhang J, Yang Y, Feng L, Kang YJ. The Association of Suppressed Hypoxia-Inducible Factor-1 Transactivation of Angiogenesis With Defective Recovery From Cerebral Ischemic Injury in Aged Rats. Front Aging Neurosci 2021; 13:648115. [PMID: 33716719 PMCID: PMC7953721 DOI: 10.3389/fnagi.2021.648115] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 02/05/2023] Open
Abstract
Elderly patients suffer more brain damage in comparison with young patients from the same ischemic stroke. The present study was undertaken to test the hypothesis that suppressed hypoxia-inducible factor-1 (HIF-1) transcription activity is responsible for defective recovery after ischemic stroke in the elders. Aged and young rats underwent 1-h transient middle cerebral artery occlusion (MCAO) to produce cerebral ischemic injury. The initial cerebral infarct volume in the young gradually declined as time elapsed, but in the aged rats remained the same. The defective recovery in the aged was associated with depressed angiogenesis and retarded neurorestoration. There was no difference in HIF-1α accumulation in the brain between the two age groups, but the expression of HIF-1 regulated genes involved in cerebral recovery was suppressed in the aged. In confirmation, inhibition of HIF-1 transactivation of gene expression in the young suppressed cerebral recovery from MCAO as the same as that observed in the aged rats. Furthermore, a copper metabolism MURR domain 1 (COMMD1) was significantly elevated after MCAO only in the brain of aged rats, and suppression of COMMD1 by siRNA targeting COMMD1 restored HIF-1 transactivation and improved recovery from MCAO-induced damage in the aged brain. These results demonstrate that impaired HIF-1 transcription activity, due at least partially to overexpression of COMMD1, is associated with the defective cerebral recovery from ischemic stroke in the aged rats.
Collapse
Affiliation(s)
- Yingjia Guo
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junpeng Zhou
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xianglong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ying Xiao
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyao Zhang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yutao Yang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Feng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Memphis Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
33
|
Cytochrome P450 CYP2E1 Suppression Ameliorates Cerebral Ischemia Reperfusion Injury. Antioxidants (Basel) 2021; 10:antiox10010052. [PMID: 33466250 PMCID: PMC7824747 DOI: 10.3390/antiox10010052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/24/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022] Open
Abstract
Despite existing strong evidence on oxidative markers overproduction following ischemia/reperfusion (I/R), the mechanism by which oxidative enzyme Cytochrome P450-2E1 (CYP2E1) contributes to I/R outcomes is not clear. In this study, we sought to evaluate the functional significance of CYP2E1 in I/R. CYP2E1 KO mice and controls were subjected to middle cerebral artery occlusion (MCAo-90 min) followed by 24 h of reperfusion to induce focal I/R injury as an acute stage model. Then, histological and chemical analyses were conducted to investigate the role of CYP2E1 in lesion volume, oxidative stress, and inflammation exacerbation. Furthermore, the role of CYP2E1 on the blood-brain barrier (BBB) integrity was investigated by measuring 20-hydroxyecosatetraenoic acid (20-HETE) activity, as well as, in vivo BBB transfer rate. Following I/R, the CYP2E1 KO mice exhibited a significantly lower lesion volume, and neurological deficits compared to controls (p < 0.005). Moreover, reactive oxygen species (ROS) production, apoptosis, and neurodegeneration were significantly lower in the CYP2E1(−/−) I/R group (p < 0.001). The BBB damage was significantly lower in CYP2E1(−/−) mice compared to wild-type (WT) (p < 0.001), while 20-HETE production was increased by 41%. Besides, inflammatory cytokines expression and the number of activated microglia were significantly lower in CYP2E1(−/−) mice following I/R. CYP2E1 suppression ameliorates I/R injury and protects BBB integrity by reducing both oxidative stress and inflammation.
Collapse
|
34
|
Liebenstund L, Coburn M, Fitzner C, Willuweit A, Langen KJ, Liu J, Veldeman M, Höllig A. Predicting experimental success: a retrospective case-control study using the rat intraluminal thread model of stroke. Dis Model Mech 2020; 13:dmm044651. [PMID: 33093066 PMCID: PMC7790196 DOI: 10.1242/dmm.044651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 10/13/2020] [Indexed: 12/23/2022] Open
Abstract
The poor translational success rate of preclinical stroke research may partly be due to inaccurate modelling of the disease. We provide data on transient middle cerebral artery occlusion (tMCAO) experiments, including detailed intraoperative monitoring to elaborate predictors indicating experimental success (ischemia without occurrence of confounding pathologies). The tMCAO monitoring data (bilateral cerebral blood flow, CBF; heart rate, HR; and mean arterial pressure, MAP) of 16 animals with an 'ideal' outcome (MCA-ischemia), and 48 animals with additional or other pathologies (subdural haematoma or subarachnoid haemorrhage), were checked for their prognostic performance (receiver operating characteristic curve and area under the curve, AUC). Animals showing a decrease in the contralateral CBF at the time of MCA occlusion suffered from unintended pathologies. Implementation of baseline MAP, in addition to baseline HR (AUC, 0.83, 95% c.i. 0.68 to 0.97), increased prognostic relevance (AUC, 0.89, 95% c.i. 0.79 to 0.98). Prediction performance improved when two additional predictors referring to differences in left and right CBF were considered (AUC, 1.00, 95% c.i. 1.0 to 1.0). Our data underline the importance of peri-interventional monitoring to verify a successful experimental performance in order to ensure a disease model as homogeneous as possible.
Collapse
Affiliation(s)
- Lisa Liebenstund
- Department of Anesthesiology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, D-52074 Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, D-52074 Aachen, Germany
| | - Christina Fitzner
- Department of Anesthesiology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, D-52074 Aachen, Germany
- 3CARE, Cardiovascular Critical Care & Anesthesia Research, University Hospital Aachen, RWTH Aachen University, D-52047 Aachen, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich GmbH, D-52428 Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Forschungszentrum Jülich GmbH, D-52428 Jülich, Germany
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, D-52047 Aachen, Germany
| | - Jingjin Liu
- Department of Anesthesiology, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, D-52074 Aachen, Germany
| | - Michael Veldeman
- Department of Neurosurgery, University Hospital Aachen, RWTH Aachen University, D-52047 Aachen, Germany
| | - Anke Höllig
- Department of Neurosurgery, University Hospital Aachen, RWTH Aachen University, D-52047 Aachen, Germany
| |
Collapse
|
35
|
Human placental mesenchymal stem cells improve stroke outcomes via extracellular vesicles-mediated preservation of cerebral blood flow. EBioMedicine 2020; 63:103161. [PMID: 33348090 PMCID: PMC7753936 DOI: 10.1016/j.ebiom.2020.103161] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/30/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Background Besides long-term trans-differentiation into neural cells, benefits of stem cell therapy (SCT) in ischemic stroke may include secretion of protective factors, which partly reflects extracellular vesicle (EVs) released by stem cell. However, the mechanism(s) by which stem cells/EVs limit stroke injury have yet to be fully defined. Methods We evaluated the protection effect of human placenta mesenchymal stem cells (hPMSC) as a potential form of SCT in experimental ischemic stroke ‘transient middle cerebral artery occusion (MCAO)/reperfusion’ mice model. Findings We found for the first time that intraperitoneal administration of hPMSCs or intravenous hPMSC-derived EVs, given at the time of reperfusion, significantly protected the ipsilateral hemisphere from ischemic injury. This protection was associated with significant restoration of normal blood flow to the post-MCAO brain. More importantly, EVs derived from hPMSC promote paracrine-based protection of SCT in the MCAO model in a cholesterol/lipid-dependent manner. Interpretation Together, our results demonstrated beneficial effects of hPMSC/EVs in experimental stroke models which could permit the rapid “translation” of these cells into clinical trials in the near-term.
Collapse
|
36
|
Early Electroacupuncture Extends the rtPA Time Window to 6 h in a Male Rat Model of Embolic Stroke via the ERK1/2-MMP9 Pathway. Neural Plast 2020. [DOI: 10.1155/2020/8851089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background. Recombinant tissue plasminogen activator (rtPA) is the only recommended pharmacological treatment for acute ischemic stroke, but it has a restricted therapeutic time window. When administered at time points greater than 4.5 h after stroke onset, rtPA disrupts the blood-brain barrier (BBB), which leads to serious brain edema and hemorrhagic transformation. Electroacupuncture (EA) exerts a neuroprotective effect on cerebral ischemia; however, researchers have not clearly determined whether EA increases the safety of thrombolysis and extends the therapeutic time window of rtPA administration following ischemic stroke. Objective. The present study was conducted to test the hypothesis that EA extends the therapeutic time window of rtPA for ischemic stroke in a male rat model of embolic stroke. Methods. SD rats were randomly divided into the sham operation group, model group, rtPA group, EA+rtPA group, and rtPA+MEK1/2 inhibitor group. An injection of rtPA was administered 6 h after ischemia. Rats were treated with EA at the Shuigou (GV26) and Neiguan (PC6) acupoints at 2 h after ischemia. Neurological function, infarct volume, BBB permeability, brain edema, and hemorrhagic transformation were assessed at 24 h after ischemia. Western blotting and immunofluorescence staining were performed to detect the levels of proteins involved in the ERK1/2 signaling pathway (MEK1/2 and ERK1/2), tight junction proteins (Claudin5 and ZO-1), and MMP9 in the ischemic penumbra at 24 h after stroke. Results. Delayed rtPA treatment aggravated hemorrhagic transformation and brain edema. However, treatment with EA plus rtPA significantly improved neurological function and reduced the infarct volume, hemorrhagic transformation, brain edema, and EB leakage in rats compared with rtPA alone. EA increased the levels of tight junction proteins, inhibited the activation of the ERK1/2 signaling pathway, and reduced MMP9 overexpression induced by delayed rtPA thrombolysis. Conclusions. EA potentially represents an effective adjunct method to increase the safety of thrombolytic therapy and extend the therapeutic time window of rtPA administration following ischemic stroke. This neuroprotective effect may be mediated by the inhibition of the ERK1/2-MMP9 pathway and alleviation of the destruction of the BBB.
Collapse
|
37
|
Liu C, Xie J, Sun S, Li H, Li T, Jiang C, Chen X, Wang J, Le A, Wang J, Li Z, Wang J, Wang W. Hemorrhagic Transformation After Tissue Plasminogen Activator Treatment in Acute Ischemic Stroke. Cell Mol Neurobiol 2020; 42:621-646. [PMID: 33125600 DOI: 10.1007/s10571-020-00985-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022]
Abstract
Hemorrhagic transformation (HT) is a common complication after thrombolysis with recombinant tissue-type plasminogen activator (rt-PA) in ischemic stroke. In this article, recent research progress of HT in vivo and in vitro studies was reviewed. We have discussed new potential mechanisms and possible experimental models of HT development, as well as possible biomarkers and treatment methods. Meanwhile, we compared and analyzed rodent models, large animal models and in vitro BBB models of HT, and the limitations of these models were discussed. The molecular mechanism of HT was investigated in terms of BBB disruption, rt-PA neurotoxicity and the effect of neuroinflammation, matrix metalloproteinases, reactive oxygen species. The clinical features to predict HT were represented including blood biomarkers and clinical factors. Recent progress in neuroprotective strategies to improve HT after stroke treated with rt-PA is outlined. Further efforts need to be made to reduce the risk of HT after rt-PA therapy and improve the clinical prognosis of patients with ischemic stroke.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jie Xie
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Shanshan Sun
- Department of Ultrasound Imaging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Hui Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Tianyu Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China
| | - Junmin Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China
| | - Anh Le
- Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Jiarui Wang
- The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhanfei Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jian Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China.
| | - Wei Wang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
38
|
Kuriakose D, Xiao Z. Pathophysiology and Treatment of Stroke: Present Status and Future Perspectives. Int J Mol Sci 2020; 21:E7609. [PMID: 33076218 PMCID: PMC7589849 DOI: 10.3390/ijms21207609] [Citation(s) in RCA: 523] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is the second leading cause of death and a major contributor to disability worldwide. The prevalence of stroke is highest in developing countries, with ischemic stroke being the most common type. Considerable progress has been made in our understanding of the pathophysiology of stroke and the underlying mechanisms leading to ischemic insult. Stroke therapy primarily focuses on restoring blood flow to the brain and treating stroke-induced neurological damage. Lack of success in recent clinical trials has led to significant refinement of animal models, focus-driven study design and use of new technologies in stroke research. Simultaneously, despite progress in stroke management, post-stroke care exerts a substantial impact on families, the healthcare system and the economy. Improvements in pre-clinical and clinical care are likely to underpin successful stroke treatment, recovery, rehabilitation and prevention. In this review, we focus on the pathophysiology of stroke, major advances in the identification of therapeutic targets and recent trends in stroke research.
Collapse
Affiliation(s)
| | - Zhicheng Xiao
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia;
| |
Collapse
|
39
|
Ma R, Xie Q, Li Y, Chen Z, Ren M, Chen H, Li H, Li J, Wang J. Animal models of cerebral ischemia: A review. Biomed Pharmacother 2020; 131:110686. [PMID: 32937247 DOI: 10.1016/j.biopha.2020.110686] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/09/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Stroke seriously threatens human health because of its characteristics of high morbidity, disability, recurrence, and mortality, thus representing a heavy financial and mental burden to affected families and society. Many preclinical effective drugs end in clinical-translation failure. Animal models are an important approach for studying diseases and drug effects, and play a central role in biomedical research. Some details about animal models of cerebral ischemia have not been published, such as left-/right-sided lesions or permanent cerebral ischemia/cerebral ischemia-reperfusion. In this review, ischemia in the left- and right-hemisphere in patients with clinical stroke and preclinical studies were compared for the first time, as were the mechanisms of permanent cerebral ischemia and cerebral ischemia-reperfusion in different phases of the disease. The results showed that stroke in the left hemisphere was more common in clinical patients, and that most patients with stroke failed to achieve successful recanalization. Significant differences were detected between permanent cerebral ischemia and cerebral ischemia-reperfusion models in the early, subacute, and recovery phases. Therefore, it is recommended that, with the exception of the determined experimental purpose or drug mechanism, left-sided permanent cerebral ischemia animal models should be prioritized, as they would be more in line with the clinical scenario and would promote clinical translation. In addition, other details regarding the preoperative management, surgical procedures, and postoperative care of these animals are provided, to help establish a precise, effective, and reproducible model of cerebral ischemia model and establish a reference for researchers in this field.
Collapse
Affiliation(s)
- Rong Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Xie
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhuoping Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mihong Ren
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hai Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hongyan Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinxiu Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
40
|
Bobot M, Thomas L, Moyon A, Fernandez S, McKay N, Balasse L, Garrigue P, Brige P, Chopinet S, Poitevin S, Cérini C, Brunet P, Dignat-George F, Burtey S, Guillet B, Hache G. Uremic Toxic Blood-Brain Barrier Disruption Mediated by AhR Activation Leads to Cognitive Impairment during Experimental Renal Dysfunction. J Am Soc Nephrol 2020; 31:1509-1521. [PMID: 32527975 DOI: 10.1681/asn.2019070728] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Uremic toxicity may play a role in the elevated risk of developing cognitive impairment found among patients with CKD. Some uremic toxins, like indoxyl sulfate, are agonists of the transcription factor aryl hydrocarbon receptor (AhR), which is widely expressed in the central nervous system and which we previously identified as the receptor of indoxyl sulfate in endothelial cells. METHODS To characterize involvement of uremic toxins in cerebral and neurobehavioral abnormalities in three rat models of CKD, we induced CKD in rats by an adenine-rich diet or by 5/6 nephrectomy; we also used AhR-/- knockout mice overloaded with indoxyl sulfate in drinking water. We assessed neurologic deficits by neurobehavioral tests and blood-brain barrier disruption by SPECT/CT imaging after injection of 99mTc-DTPA, an imaging marker of blood-brain barrier permeability. RESULTS In CKD rats, we found cognitive impairment in the novel object recognition test, the object location task, and social memory tests and an increase of blood-brain barrier permeability associated with renal dysfunction. We found a significant correlation between 99mTc-DTPA content in brain and both the discrimination index in the novel object recognition test and indoxyl sulfate concentrations in serum. When we added indoxyl sulfate to the drinking water of rats fed an adenine-rich diet, we found an increase in indoxyl sulfate concentrations in serum associated with a stronger impairment in cognition and a higher permeability of the blood-brain barrier. In addition, non-CKD AhR-/- knockout mice were protected against indoxyl sulfate-induced blood-brain barrier disruption and cognitive impairment. CONCLUSIONS AhR activation by indoxyl sulfate, a uremic toxin, leads to blood-brain barrier disruption associated with cognitive impairment in animal models of CKD.
Collapse
Affiliation(s)
- Mickaël Bobot
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, Assistnce Publique - Hôpitaux de Marseille, Marseille, France .,Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Laurent Thomas
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Anaïs Moyon
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France.,Service de Radiopharmacie, Assistnce Publique - Hôpitaux de Marseille, Marseille, France
| | - Samantha Fernandez
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | - Nathalie McKay
- Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Laure Balasse
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France
| | - Philippe Garrigue
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France.,Service de Radiopharmacie, Assistnce Publique - Hôpitaux de Marseille, Marseille, France
| | - Pauline Brige
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Laboratoire d'Imagerie Interventionelle Expérimentale, Aix-Marseille Université, Marseille, France
| | - Sophie Chopinet
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Laboratoire d'Imagerie Interventionelle Expérimentale, Aix-Marseille Université, Marseille, France.,Service de Chirurgie générale et transplantation hépatique, Hôpital de la Timone, Assistnce Publique - Hôpitaux de Marseille, Marseille, France
| | - Stéphane Poitevin
- Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Claire Cérini
- Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Philippe Brunet
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, Assistnce Publique - Hôpitaux de Marseille, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Françoise Dignat-George
- Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Stéphane Burtey
- Centre de Néphrologie et Transplantation Rénale, Hôpital de la Conception, Assistnce Publique - Hôpitaux de Marseille, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France
| | - Benjamin Guillet
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France.,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France.,Service de Radiopharmacie, Assistnce Publique - Hôpitaux de Marseille, Marseille, France
| | - Guillaume Hache
- Centre Européen de recherche en Imagerie Médicale, Aix Marseille Université, Centre National de la Recherche Scientifique, Marseille, France .,Centre de Recherche en Cardiovasculaireet Nutrition, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut National de Recherche pour l'agriculture, l'alimentation et l'environnement, Marseille, France.,Pharmacie, Hôpital de la Timone, Assistnce Publique - Hôpitaux de Marseille, Marseille, France
| |
Collapse
|
41
|
Vollert J, Schenker E, Macleod M, Bespalov A, Wuerbel H, Michel M, Dirnagl U, Potschka H, Waldron AM, Wever K, Steckler T, van de Casteele T, Altevogt B, Sil A, Rice ASC. Systematic review of guidelines for internal validity in the design, conduct and analysis of preclinical biomedical experiments involving laboratory animals. BMJ OPEN SCIENCE 2020; 4:e100046. [PMID: 35047688 PMCID: PMC8647591 DOI: 10.1136/bmjos-2019-100046] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/10/2019] [Accepted: 01/15/2020] [Indexed: 02/01/2023] Open
Abstract
Over the last two decades, awareness of the negative repercussions of flaws in the planning, conduct and reporting of preclinical research involving experimental animals has been growing. Several initiatives have set out to increase transparency and internal validity of preclinical studies, mostly publishing expert consensus and experience. While many of the points raised in these various guidelines are identical or similar, they differ in detail and rigour. Most of them focus on reporting, only few of them cover the planning and conduct of studies. The aim of this systematic review is to identify existing experimental design, conduct, analysis and reporting guidelines relating to preclinical animal research. A systematic search in PubMed, Embase and Web of Science retrieved 13 863 unique results. After screening these on title and abstract, 613 papers entered the full-text assessment stage, from which 60 papers were retained. From these, we extracted unique 58 recommendations on the planning, conduct and reporting of preclinical animal studies. Sample size calculations, adequate statistical methods, concealed and randomised allocation of animals to treatment, blinded outcome assessment and recording of animal flow through the experiment were recommended in more than half of the publications. While we consider these recommendations to be valuable, there is a striking lack of experimental evidence on their importance and relative effect on experiments and effect sizes.
Collapse
Affiliation(s)
- Jan Vollert
- Pain Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Esther Schenker
- Institut de Recherches Internationales Servier, Suresnes, Île-de-France, France
| | - Malcolm Macleod
- Centre for Clinical Brain Sciences, Edinburgh Medical School, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Anton Bespalov
- Partnership for Assessment and Accreditation of Scientific Practice, Heidelberg, Germany
- Valdman Institute of Pharmacology, Pavlov First State Medical University of Saint Petersburg, Sankt Petersburg, Russian Federation
| | - Hanno Wuerbel
- Division of Animal Welfare, Vetsuisse Faculty, VPH Institute, University of Bern, Bern, Switzerland
| | - Martin Michel
- Universitätsmedizin Mainz, Johannes Gutenberg Universität Mainz, Mainz, Rheinland-Pfalz, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universitat Munchen, Munchen, Bayern, Germany
| | - Ann-Marie Waldron
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universitat Munchen, Munchen, Bayern, Germany
| | - Kimberley Wever
- Systematic Review Centre for Laboratory Animal Experimentation, Department for Health Evidence, Nijmegen Institute for Health Sciences, Radboud Universiteit, Nijmegen, Gelderland, Netherlands
| | | | | | | | - Annesha Sil
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Andrew S C Rice
- Pain Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
42
|
Abstract
Despite thousands of neuroprotectants demonstrating promise in preclinical trials, a neuroprotective therapeutic has yet to be approved for the treatment of acute brain injuries such as stroke or traumatic brain injury. Developing a more detailed understanding of models and populations demonstrating "neurological resilience" in spite of brain injury can give us important insights into new translational therapies. Resilience is the process of active adaptation to a stressor. In the context of neuroprotection, models of preconditioning and unique animal models of extreme physiology (such as hibernating species) reliably demonstrate resilience in the laboratory setting. In the clinical setting, resilience is observed in young patients and can be found in those with specific genetic polymorphisms. These important examples of resilience can help transform and extend the current neuroprotective framework from simply countering the injurious cascade into one that anticipates, monitors, and optimizes patients' physiological responses from the time of injury throughout the process of recovery. This review summarizes the underpinnings of key adaptations common to models of resilience and how this understanding can be applied to new neuroprotective approaches.
Collapse
Affiliation(s)
- Neel S Singhal
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA.
| | - Chung-Huan Sun
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Evan M Lee
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Dengke K Ma
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| |
Collapse
|
43
|
Wang J, Zhang P, Tang Z. Animal models of transient ischemic attack: a review. Acta Neurol Belg 2020; 120:267-275. [PMID: 32048230 PMCID: PMC7083805 DOI: 10.1007/s13760-020-01295-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/30/2020] [Indexed: 12/29/2022]
Abstract
Transient ischemic attack (TIA) is defined as a brief episode of neurological dysfunction caused by focal cerebral ischemia. TIA is a critical early warning signal of stroke. Patients with TIA may have long-term cognitive decline. The pathogenesis and pathological changes of TIA have not been fully elucidated. Animal models can simulate the process of human diseases and are essential tools to investigate injury mechanisms and therapeutic approaches of TIA. Most TIA animal models are based on ischemic stroke models and the definition of TIA. Each model has unique strengths and weaknesses. The establishment of a successful and reliable TIA model should follow three criteria: (1) objective evidence of cerebral arteries occlusion and reperfusion, (2) no permanent neurological deficit, and (3) no acute cerebral infarction. However, experimental animal models are impossible to be completely consistent with human TIA, because TIA itself is a heterogeneous disease. In the present review, the selection of animals, methodological development, and evaluation of cerebral blood flow of animal models of TIA are comprehensively evaluated.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430030 China
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430030 China
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan, 430030 China
| |
Collapse
|
44
|
Dhir N, Medhi B, Prakash A, Goyal MK, Modi M, Mohindra S. Pre-clinical to Clinical Translational Failures and Current Status of Clinical Trials in Stroke Therapy: A Brief Review. Curr Neuropharmacol 2020; 18:596-612. [PMID: 31934841 PMCID: PMC7457423 DOI: 10.2174/1570159x18666200114160844] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/31/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022] Open
Abstract
In stroke (cerebral ischemia), despite continuous efforts both at the experimental and clinical level, the only approved pharmacological treatment has been restricted to tissue plasminogen activator (tPA). Stroke is the leading cause of functional disability and mortality throughout worldwide. Its pathophysiology starts with energy pump failure, followed by complex signaling cascade that ultimately ends in neuronal cell death. Ischemic cascade involves excessive glutamate release followed by raised intracellular sodium and calcium influx along with free radicals' generation, activation of inflammatory cytokines, NO synthases, lipases, endonucleases and other apoptotic pathways leading to cell edema and death. At the pre-clinical stage, several agents have been tried and proven as an effective neuroprotectant in animal models of ischemia. However, these agents failed to show convincing results in terms of efficacy and safety when the trials were conducted in humans following stroke. This article highlights the various agents which have been tried in the past but failed to translate into stroke therapy along with key points that are responsible for the lagging of experimental success to translational failure in stroke treatment.
Collapse
Affiliation(s)
| | - Bikash Medhi
- Address correspondence to this author at the Department of Pharmacology, Research Block B, 4th Floor, Room no 4043, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India; E-mail:
| | | | | | | | | |
Collapse
|
45
|
Kamarudin SN, Iezhitsa I, Tripathy M, Alyautdin R, Ismail NM. Neuroprotective effect of poly(lactic-co-glycolic acid) nanoparticle-bound brain-derived neurotrophic factor in a permanent middle cerebral artery occlusion model of ischemia in rats. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
46
|
Abstract
Novel therapeutic intervention that aims to enhance the endogenous recovery potential of the brain during the subacute phase of stroke has produced promising results. The paradigm shift in treatment approaches presents new challenges to preclinical and clinical researchers alike, especially in the functional endpoints domain. Shortcomings of the "neuroprotection" era of stroke research are yet to be fully addressed. Proportional recovery observed in clinics, and potentially in animal models, requires a thorough reevaluation of the methods used to assess recovery. To this end, this review aims to give a detailed evaluation of functional outcome measures used in clinics and preclinical studies. Impairments observed in clinics and animal models will be discussed from a functional testing perspective. Approaches needed to bridge the gap between clinical and preclinical research, along with potential means to measure the moving target recovery, will be discussed. Concepts such as true recovery of function and compensation and methods that are suitable for distinguishing the two are examined. Often-neglected outcomes of stroke, such as emotional disturbances, are discussed to draw attention to the need for further research in this area.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Burke Neurological Research Institute, White Plains, NY, USA
| | - Sunghee Cho
- Burke Neurological Research Institute, White Plains, NY, USA.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine at Burke Neurological Research Institute, White Plains, NY, USA
| |
Collapse
|
47
|
Kuts R, Frank D, Gruenbaum BF, Grinshpun J, Melamed I, Knyazer B, Tarabrin O, Zvenigorodsky V, Shelef I, Zlotnik A, Boyko M. A Novel Method for Assessing Cerebral Edema, Infarcted Zone and Blood-Brain Barrier Breakdown in a Single Post-stroke Rodent Brain. Front Neurosci 2019; 13:1105. [PMID: 31680838 PMCID: PMC6805703 DOI: 10.3389/fnins.2019.01105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/01/2019] [Indexed: 11/13/2022] Open
Abstract
Stroke is a major cause of global morbidity and mortality. Middle cerebral artery occlusion (MCAO) has historically been the most common animal model of simulating ischemic stroke. The extent of neurological injury after MCAO is typically measured by cerebral edema, infarct zone, and blood-brain barrier (BBB) permeability. A significant limitation of these methods is that separate sets of brains must be used for each measurement. Here we examine an alternative method of measuring cerebral edema, infarct zone and BBB permeability following MCAO in the same set of brain samples. Ninety-six rats were randomly divided into three experimental groups. Group 1 (n = 27) was used for the evaluation of infarct zone and brain edema in rats post-MCAO (n = 17) vs. sham-operated controls (n = 10). Group 2 (n = 27) was used for the evaluation of BBB breakdown in rats post-MCAO (n = 15) vs. sham-operated controls (n = 10). In Group 3 (n = 42), all three parameters were measured in the same set of brain slices in rats post-MCAO (n = 26) vs. sham-operated controls (n = 16). The effect of Evans blue on the accuracy of measuring infarct zone by 2,3,5-triphenyltetrazolium chloride (TTC) staining was determined by measuring infarct zone with and without an applied blue filter. The effects of various concentrations of TTC (0, 0.05, 0.35, 0.5, 1, and 2%) on the accuracy of measuring BBB permeability was also assessed. There was an increase in infarct volume (p < 0.01), brain edema (p < 0.01) and BBB breakdown (p < 0.01) in rats following MCAO compared to sham-operated controls, whether measured separately or together in the same set of brain samples. Evans blue had an effect on measuring infarct volume that was minimized by the application of a blue filter on scanned brain slices. There was no difference in the Evans blue extravasation index for the brain tissue samples without TTC compared to brain tissue samples incubated in TTC. Our results demonstrate that measuring cerebral edema, infarct zone and BBB permeability following MCAO can accurately be measured in the same set of brain samples.
Collapse
Affiliation(s)
- Ruslan Kuts
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Dmitry Frank
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Benjamin F Gruenbaum
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, United States
| | - Julia Grinshpun
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Israel Melamed
- Department of Neurosurgery, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Boris Knyazer
- Department of Ophthalmology, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Oleg Tarabrin
- Department of Anesthesiology and Intensive Care with Postgraduate Education, Odessa National Medical University, Odessa, Ukraine
| | - Vladislav Zvenigorodsky
- Department of Radiology, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ilan Shelef
- Department of Radiology, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Alexander Zlotnik
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Matthew Boyko
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
48
|
Zeb A, Cha JH, Noh AR, Qureshi OS, Kim KW, Choe YH, Shin D, Shah FA, Majid A, Bae ON, Kim JK. Neuroprotective effects of carnosine-loaded elastic liposomes in cerebral ischemia rat model. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00462-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
49
|
Chen M, Lyu H, Li T, Su XW, Leung CK, Xiong MZQ, Poon WS, Cai YF, Lu G, Chan WY, Wang LX. Study of the association between gait variability and gene expressions in a mouse model of transient focal ischemic stroke. Int J Neurosci 2019; 130:52-63. [PMID: 31512542 DOI: 10.1080/00207454.2019.1663188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Purpose: Gait variability analysis has been clinically adopted to characterize the presentation of various neurological diseases. However, literature and practice lack a comprehensive murine model assessment of the gait deficits that result from transient focal ischemic stroke. Further, correlations between gait parameters and the gene expression profiles associated with brain ischemia have yet to be identified. This study quantitatively assesses gait deficits through a murine model of transient focal cerebral ischemia on day 7 to determine associations between gait deficits and ischemia-related gene expressions.Methods: A total of 182 dynamic and static gait parameters from the transient middle cerebral artery occlusion (MCAO) murine model for simulating human transient focal ischemic stroke on day 7 were measured using the CatWalk system. Pearson's correlation analysis and genes associated with ischemia were identified from the existing literature to aid the investigation of the relationship between gait variability and gene expression profiles.Results: Thirty-nine gait parameters and the mRNA expression levels of four of the eight ischemia-associated genes exhibited more significant change in the MCAO models (p < 0.005) on day 7. Twenty-six gait parameters exhibited strong correlations with four ischemia-associated genes.Conclusion: This examination of gait variability and the strong correlation to the gene expression profiles associated with transient focal brain ischemia on day 7 provides a quantitative and reliable assessment of the MCAO model's motor performance. This research provides valuable insights into the study of disease progression and offers novel therapeutic interventions in the murine modeling of ischemic stroke.
Collapse
Affiliation(s)
- Mei Chen
- Neurology Department, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guang Zhou, China.,CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hao Lyu
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Tu Li
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xian Wei Su
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Kwan Leung
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Mark Zhi Qiang Xiong
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye-Feng Cai
- Neurology Department, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guang Zhou, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Ningxia Human Stem Cell Institute, The General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wai-Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Li-Xin Wang
- Neurology Department, The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guang Zhou, China
| |
Collapse
|
50
|
Provencher S, Archer SL, Ramirez FD, Hibbert B, Paulin R, Boucherat O, Lacasse Y, Bonnet S. Standards and Methodological Rigor in Pulmonary Arterial Hypertension Preclinical and Translational Research. Circ Res 2019; 122:1021-1032. [PMID: 29599278 DOI: 10.1161/circresaha.117.312579] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite advances in our understanding of the pathophysiology and the management of pulmonary arterial hypertension (PAH), significant therapeutic gaps remain for this devastating disease. Yet, few innovative therapies beyond the traditional pathways of endothelial dysfunction have reached clinical trial phases in PAH. Although there are inherent limitations of the currently available models of PAH, the leaky pipeline of innovative therapies relates, in part, to flawed preclinical research methodology, including lack of rigour in trial design, incomplete invasive hemodynamic assessment, and lack of careful translational studies that replicate randomized controlled trials in humans with attention to adverse effects and benefits. Rigorous methodology should include the use of prespecified eligibility criteria, sample sizes that permit valid statistical analysis, randomization, blinded assessment of standardized outcomes, and transparent reporting of results. Better design and implementation of preclinical studies can minimize inherent flaws in the models of PAH, reduce the risk of bias, and enhance external validity and our ability to distinguish truly promising therapies form many false-positive or overstated leads. Ideally, preclinical studies should use advanced imaging, study several preclinical pulmonary hypertension models, or correlate rodent and human findings and consider the fate of the right ventricle, which is the major determinant of prognosis in human PAH. Although these principles are widely endorsed, empirical evidence suggests that such rigor is often lacking in pulmonary hypertension preclinical research. The present article discusses the pitfalls in the design of preclinical pulmonary hypertension trials and discusses opportunities to create preclinical trials with improved predictive value in guiding early-phase drug development in patients with PAH, which will need support not only from researchers, peer reviewers, and editors but also from academic institutions, funding agencies, and animal ethics authorities.
Collapse
Affiliation(s)
- Steeve Provencher
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - Stephen L Archer
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - F Daniel Ramirez
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - Benjamin Hibbert
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - Roxane Paulin
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - Olivier Boucherat
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - Yves Lacasse
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada
| | - Sébastien Bonnet
- From the Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., S.B.), Institut universitaire de cardiologie et de pneumologie de Québec Research Center (S.P., R.P., O.B., Y.L., S.B.), and Department of Medicine (S.P., R.P., O.B., Y.L., S.B.), Université Laval, Québec, Canada; Department of Medicine, Queen's University, Kingston, Canada (S.L.A.); Division of Cardiology (F.D.R., B.H.), CAPITAL Research Group (F.D.R., B.H.), and Vascular Biology and Experimental Medicine Laboratory (B.H.), University of Ottawa Heart Institute, Ontario, Canada; and Department of Cellular and Molecular Medicine (B.H.) and School of Epidemiology and Public Health (F.D.R.) University of Ottawa, Ontario, Canada.
| |
Collapse
|