1
|
Sun X, Hou J, Xu H, Qu H. Efficacy of bumetanide in animal models of ischemic stroke: a systematic review and meta-analysis. Aging (Albany NY) 2024; 16:9959-9971. [PMID: 38850525 PMCID: PMC11210250 DOI: 10.18632/aging.205910] [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: 08/29/2023] [Accepted: 05/03/2024] [Indexed: 06/10/2024]
Abstract
This meta-analysis aimed to describe the efficacy of bumetanide in improving infarct volume, brain edema, and behavioral outcomes in animal models of cerebral ischemia. Embase, PubMed and Web of Science databases were searched from their inception to February 2024 (INPLASY:202430023). Data on the animal species, stroke model, drug dose, time of treatment, method of administration, study quality, and outcomes were extracted and pooled in a meta-analysis. The combined standardized mean difference (SMD) or mean difference (MD) estimates and 95% confidence intervals (CIs) were calculated using random- or fixed-effects models. Thirteen eligible studies involving >200 animals fulfilled the inclusion criteria and were included in this meta-analysis. Meta-analyses demonstrated that bumetanide treatment significantly reduced cerebral infarct volume (SMD: -0.42; 95% CI: -0.75, -0.09; p < 0.01; n = 186 animals) and consistently relieved brain edema (SMD: -1.39; 95% CI: -2.06, -0.72; p < 0.01; n = 64 animals). Subgroup analyses demonstrated that bumetanide treatment reduced infarct volume in transient but not permanent cerebral ischemia models. When administered after the stroke, it was more effective than treatment initiation before the stroke. Eight studies assessed the effect of bumetanide on behavioral function and the results showed that bumetanide treatment significantly improved neurobehavioral deficits (SMD: -2.35; 95% CI: -2.72, -1.97; p < 0.01; n = 250 animals). We conclude that bumetanide appears to be effective in reducing infarct volume and brain edema and improving behavioral recovery in animal models of cerebral ischemia. This mechanism needs to be confirmed through further investigation.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Neurology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Jiadi Hou
- Department of Neurology, The General Hospital of Northern Theater Command, Shenyang, China
| | - Haichun Xu
- Department of Psychiatry, Shenyang Jing’an Mental Health Hospital, Shenyang, China
| | - Huiling Qu
- Department of Neurology, The General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
2
|
Sarkala HB, Jahanshahi M, Dolatabadi LK, Namavar MR. G-CSF improved the memory and dendritic morphology impairments in the hippocampal CA1 pyramidal neurons after brain ischemia in the male rats. Metab Brain Dis 2023; 38:2573-2581. [PMID: 37728699 DOI: 10.1007/s11011-023-01286-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Stroke remains the leading cause of death and disability in the world. A new potential treatment for stroke is the granulocyte colony-stimulating factor (G-CSF), which exerts neuroprotective effects through multiple mechanisms. Memory impairment is the most common cognitive problem after a stroke. The suggested treatment for memory impairments is cognitive rehabilitation, which is often ineffective. The hippocampus plays an important role in memory formation. This project aimed to study the effect of G-CSF on memory and dendritic morphology of hippocampal CA1 pyramidal neurons after middle cerebral artery occlusion (MCAO)in rats. METHODS Male Sprague-Dawley rats were divided into three groups: the sham, control (MCAO + Vehicle), and treatment (MCAO + G-CSF) groups. G-CSF (50 µg/kg S.C) was administered at 6, 24, and 48 h after brain ischemia induction. The passive avoidance task to evaluate learning and memory was performed on days 6 and 7 post-ischemia. Seven days after MCAO, the brain was removed and the hippocampal slices were stained with Golgi. After that, the neurons were analyzed for dendritic morphology and maturity. OUTCOMES The data showed that stroke was associated with a significant impairment in the acquisition and retention of passive avoidance tasks, while the G-CSF improved learning and memory loss. The dendritic length, arborization, spine density, and mature spines of the hippocampus CA1 neurons were significantly reduced in the control group, and treatment with G-CSF significantly increased these parameters. CONCLUSION G-CSF, even with three doses, improved learning and memory deficits, and dendritic morphological changes in the CA1 hippocampal neurons resulted from brain ischemia.
Collapse
Affiliation(s)
- Hamzeh Badeli Sarkala
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Leila Kamali Dolatabadi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Reza Namavar
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Dharra R, Kumar Sharma A, Datta S. Emerging aspects of cytokine storm in COVID-19: The role of proinflammatory cytokines and therapeutic prospects. Cytokine 2023; 169:156287. [PMID: 37402337 PMCID: PMC10291296 DOI: 10.1016/j.cyto.2023.156287] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/24/2023] [Indexed: 07/06/2023]
Abstract
COVID-19 has claimed millions of lives during the last 3 years since initial cases were reported in Wuhan, China, in 2019. Patients with COVID-19 suffer from severe pneumonia, high fever, acute respiratory distress syndrome (ARDS), and multiple-organ dysfunction, which may also result in fatality in extreme cases. Cytokine storm (CS) is hyperactivation of the immune system, wherein the dysregulated production of proinflammatory cytokines could result in excessive immune cell infiltrations in the pulmonary tissues, resulting in tissue damage. The immune cell infiltration could also occur in other tissues and organs and result in multiple organs' dysfunction. The key cytokines implicated in the onset of disease severity include TNF-α, IFN-γ, IL-6, IL-1β, GM-CSF, and G-CSF. Controlling the CS is critical in treating COVID-19 disease. Therefore, different strategies are employed to mitigate the effects of CS. These include using monoclonal antibodies directed against soluble cytokines or the cytokine receptors, combination therapies, mesenchymal stem cell therapy, therapeutic plasma exchange, and some non-conventional treatment methods to improve patient immunity. The current review describes the role/s of critical cytokines in COVID-19-mediated CS and the respective treatment modalities.
Collapse
Affiliation(s)
- Renu Dharra
- CSIR-Institute of Microbial Technology, Sector 39 A, Chandigarh 160036, India
| | - Anil Kumar Sharma
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Sonal Datta
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| |
Collapse
|
4
|
Qureshi AI, Akhtar IN, Ma X, Lodhi A, Bhatti I, Beall J, Broderick JP, Cassarly CN, Martin RH, Sharma R, Thakkar M, Suarez JI. Effect of Cilostazol in Animal Models of Cerebral Ischemia and Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis. Neurocrit Care 2023; 38:698-713. [PMID: 36450971 DOI: 10.1007/s12028-022-01637-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/27/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Cilostazol, a phosphodiesterase III inhibitor, appears to be a promising agent for preventing cerebral ischemia in patients with aneurysmal subarachnoid hemorrhage. Here, the authors perform a systematic review and meta-analysis to quantitatively assess the effects of cilostazol on brain structural and functional outcomes in animal models of cerebral ischemia and subarachnoid hemorrhage-induced cerebral vasospasm. METHODS By using the PRISMA guidelines, a search of the PubMed, Scopus, and Web of Science was conducted to identify relevant studies. Study quality of each included study for both systematic reviews were scored by using an adapted 15-item checklist from the Collaborative Approach to Meta-Analysis of Animal Data from Experimental Studies. We calculated a standardized mean difference as effect size for each comparison. For each outcome, comparisons were combined by using random-effects modeling to account for heterogeneity, with a restricted maximum likelihood estimate of between-study variance. RESULTS A total of 22 (median [Q1, Q3] quality score of 7 [5, 8]) and 6 (median [Q1, Q3] quality score of 6 [6, 6]) studies were identified for cerebral ischemia and subarachnoid hemorrhage-induced cerebral vasospasm, respectively. Cilostazol significantly reduced the infarct volume in cerebral ischemia models with a pooled standardized mean difference estimate of - 0.88 (95% confidence interval [CI] [- 1.07 to - 0.70], p < 0.0001). Cilostazol significantly reduced neurofunctional deficits in cerebral ischemia models with a pooled standardized mean difference estimate of - 0.66 (95% CI [- 1.06 to - 0.28], p < 0.0001). Cilostazol significantly improved the basilar artery diameter in subarachnoid hemorrhage-induced cerebral vasospasm with a pooled standardized mean difference estimate of 2.30 (95% CI [0.94 to 3.67], p = 0.001). Cilostazol also significantly improved the basilar artery cross-section area with a pooled standardized mean estimate of 1.88 (95% CI [0.33 to 3.43], p < 0.05). Overall, there was between-study heterogeneity and asymmetry in the funnel plot observed in all comparisons. CONCLUSIONS Published animal data support the overall efficacy of cilostazol in reducing infarct volume and neurofunctional deficits in cerebral ischemia models and cerebral vasospasm in subarachnoid hemorrhage models.
Collapse
Affiliation(s)
- Adnan I Qureshi
- Department of Neurology, University of Missouri, Columbia, MO, USA.
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA.
| | - Iqra N Akhtar
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Xiaoyu Ma
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Abdullah Lodhi
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Ibrahim Bhatti
- Department of Neurology, University of Missouri, Columbia, MO, USA
- Zeenat Qureshi Stroke Institute, St. Cloud, MN, USA
| | - Jonathan Beall
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | | | - Christy N Cassarly
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Renee H Martin
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Rishi Sharma
- Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Mahesh Thakkar
- Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Jose I Suarez
- Division of Neurosciences Critical Care, Departments of Anesthesiology and Critical Care Medicine, Neurology, and Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Singh D, Wasan H, Reeta KH. Preclinical Stroke Research and Translational Failure: A Bird's Eye View on Preventable Variables. Cell Mol Neurobiol 2022; 42:2003-2017. [PMID: 33786698 PMCID: PMC11421600 DOI: 10.1007/s10571-021-01083-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
6
|
Zheng Y, Zhou J, Wang Y, Fan F, Liu S, Wang Y. Neural Stem/Progenitor Cell Transplantation in Parkinson's Rodent Animals: A Meta-Analysis and Systematic Review. Stem Cells Transl Med 2022; 11:383-393. [PMID: 35325234 PMCID: PMC9052406 DOI: 10.1093/stcltm/szac006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 01/08/2022] [Indexed: 12/02/2022] Open
Abstract
The effects of neural stem/progenitor cells (NSPCs) have been extensively evaluated by multiple studies in animal models of Parkinson's disease (PD), but the therapeutic efficacy was inconsistent. Here, we searched 4 databases (PubMed, Embase, Scopus, and Web of Science) and performed a meta-analysis to estimate the therapeutic effects of unmodified NSPCs on neurological deficits in rodent animal models of PD. Data on study quality score, behavioral outcomes (apomorphine or amphetamine-induced rotation and limb function), histological outcome (densitometry of TH+ staining in the SNpc), and cell therapy-related severe adverse events were extracted for meta-analysis and systematic review. Twenty-one studies with a median quality score of 6 (range from 4 to 9) in 11 were examined. Significant improvement was observed in the overall pooled standardized mean difference (SMD) between animals transplanted with NSPCs and with control medium (1.22 for apomorphine-induced rotation, P < .001; 1.50 for amphetamine-induced rotation, P < .001; 0.86 for limb function, P < .001; and -1.96 for the densitometry of TH+ staining, P < .001). Further subgroup analysis, animal gender, NSPCs source, NSPCs dosage, and pretreatment behavioral assessment were closely correlated with apomorphine-induced rotation and amphetamine-induced rotation. In conclusion, unmodified NSPCs therapy attenuated behavioral deficits and increased dopaminergic neurons in rodent PD models, supporting the consideration of early-stage clinical trial of NSPCs in patients with PD.
Collapse
Affiliation(s)
- Yifeng Zheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jun Zhou
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yisai Wang
- Department of Electrical and Computer Engineering, Rutgers University-New Brunswick, New Jersey, USA
| | - Fanfan Fan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yu Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
7
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
8
|
Nistal D, Ali M, Wei D, Mocco J, Kellner C. A Systematic Review and Meta-Analysis of Statins in Animal Models of Intracerebral Hemorrhage. World Neurosurg 2021; 155:32-40. [PMID: 34384917 DOI: 10.1016/j.wneu.2021.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a severe form of stroke with limited treatment options. Statins have shown promise as a therapy for ICH in animal and human studies. We systematically reviewed and assessed the quality of preclinical studies exploring statin-use after ICH to guide clinical trial decision-making and design. METHODS We identified preclinical trials assessing the efficacy of statins in ICH via a systematic review of the literature according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines. In total, 16 studies were identified that described statin use in an animal model of ICH and assessed histological outcomes, behavioral scores, or both. Design characteristics were analyzed using Stroke Therapy Academic Industry Roundtable (STAIR) criteria modified for ICH. Meta-analysis was performed using a random effects model. RESULTS Behavioral outcomes were assessed in 12 of the studies with 100% (n = 12) reporting that statins significantly improved ICH recovery. Histologic hematoma volume and brain water content outcomes were analyzed in 10 of the studies, with 50% (n = 5) reporting significant improvement. The ratio of means between experimental and control cases for modified Neurological Severity Score was 0.63 (95% confidence interval 0.49-0.82). The ratio of means between experimental and control cases for hemorrhagic volume was 0.85 (95% confidence interval 0.70-1.03). There was heterogeneity between studies (P < 0.0001) but no evidence of publication bias (P = 0.89, P = 0.59, respectively). CONCLUSIONS Behavioral outcomes in ICH were found to consistently improve with administration of statins in preclinical studies suggesting that statin therapy may be suitable for randomized clinical trials in humans. In addition, the STAIR criteria can be modified to effectively evaluate preclinical studies in ICH.
Collapse
Affiliation(s)
- Dominic Nistal
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Muhammad Ali
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| | - Daniel Wei
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - J Mocco
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christopher Kellner
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
9
|
Wall A, Anger O, Jood K, Blomstrand C, Andreasson U, Blennow K, Zetterberg H, Isgaard J, Jern C, Åberg ND, Svensson J. Circulating granulocyte colony-stimulating factor and functional outcome after ischemic stroke: an observational study. Neurol Res 2021; 43:1013-1022. [PMID: 34253146 DOI: 10.1080/01616412.2021.1948766] [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: 10/20/2022]
Abstract
Objectives: While granulocyte colony-stimulating factor (G-CSF) has shown beneficial effects in experimental ischemic stroke (IS), these effects have not been reproduced clinically. Small-to-medium-sized observational studies have reported varying associations for G-CSF with stroke severity and post-stroke functional outcome, prompting their investigation in a larger study.Methods: Endogenous serum G-CSF (S-GCSF) was measured in the acute phase and after 3 months in patients with IS (N = 435; 36% females; mean age, 57 years) from the Sahlgrenska Academy Study on Ischemic Stroke (SAHLSIS). Stroke severity was scored according to the National Institutes of Health Stroke Scale (NIHSS), and the modified Rankin Scale (mRS) assessed functional outcomes at 3-month and 2-year post-stroke. Correlation and logistic regression analyses with confounder adjustments assessed the relationships.Results: The acute S-GCSF level was 23% higher than at 3-month post-stroke (p < 0.001). Acute G-CSF correlated weakly with stroke severity quintiles (r = 0.12, p = 0.013) and with high-sensitivity C-reactive protein (r = 0.29, p < 0.001). The association between S-GCSF (as quintiles, q) and poor functional outcome at 3 months (mRS 3-6; S-GCSF-q5 vs. S-GCSF-q1, age- and sex-adjusted odds ratio: 4.27, 95% confidence interval: 1.82-9.99; p = 0.001) withstood adjustment for cardiovascular risk factors and stroke subtype, but not additional correction for stroke severity. Post-stroke changes in S-GSCF and absolute 3-month S-GCSF were not associated with 3-month or 2-year functional outcomes.Discussion: Early post-stroke S-GCSF is increased in severe IS and associated with 3-month poor functional outcomes. The change in S-GCSF and the 3-month S-GCSF appear to be less-important, and S-GCSF likely reflects inflammation in large infarctions.
Collapse
Affiliation(s)
- Alexander Wall
- Department of Internal Medicine, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Department of Acute Medicine and Geriatrics (Su/Sahlgrenska), Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Olof Anger
- Department of Internal Medicine, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Katarina Jood
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Christian Blomstrand
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Stroke Centre West, Sahlgrenska Academy at University of Gothenburg, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, UCL, Gower St., London, UK
| | - Jörgen Isgaard
- Department of Internal Medicine, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Department of Acute Medicine and Geriatrics (Su/Sahlgrenska), Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Christina Jern
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Department of Clinical Pathology and Genetics, Institute of Biomedicine, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - N David Åberg
- Department of Internal Medicine, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Sweden.,Department of Acute Medicine and Geriatrics (Su/Sahlgrenska), Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal Medicine, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| |
Collapse
|
10
|
Narayan SK, Grace Cherian S, Babu Phaniti P, Babu Chidambaram S, Rachel Vasanthi AH, Arumugam M. Preclinical animal studies in ischemic stroke: Challenges and some solutions. Animal Model Exp Med 2021; 4:104-115. [PMID: 34179718 PMCID: PMC8212819 DOI: 10.1002/ame2.12166] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 01/28/2021] [Indexed: 01/01/2023] Open
Abstract
Despite the impressive efficacies demonstrated in preclinical research, hundreds of potentially neuroprotective drugs have failed to provide effective neuroprotection for ischemic stroke in human clinical trials. Lack of a powerful animal model for human ischemic stroke could be a major reason for the failure to develop successful neuroprotective drugs for ischemic stroke. This review recapitulates the available cerebral ischemia animal models, provides an anatomical comparison of the circle of Willis of each species, and describes the functional assessment tests used in these ischemic stroke models. The distinct differences between human ischemic stroke and experimental stroke in available animal models is explored. Innovative animal models more closely resembling human strokes, better techniques in functional outcome assessment and better experimental designs generating clearer and stronger evidence may help realise the development of truly neuroprotective drugs that will benefit human ischemic stroke patients. This may involve use of newer molecules or revisiting earlier studies with new experimental designs. Translation of any resultant successes may then be tested in human clinical trials with greater confidence and optimism.
Collapse
Affiliation(s)
- Sunil K. Narayan
- Comprehensive Stroke Care and Neurobiology Centre, Department of NeurologyJawaharlal Institute of Postgraduate Medical Education and ResearchPuducherryIndia
| | - Simy Grace Cherian
- Comprehensive Stroke Care and Neurobiology Centre, Department of NeurologyJawaharlal Institute of Postgraduate Medical Education and ResearchPuducherryIndia
| | - Prakash Babu Phaniti
- Department of Biotechnology & School of Medical SciencesUniversity of HyderabadHyderabadIndia
| | | | | | - Murugesan Arumugam
- Comprehensive Stroke Care and Neurobiology Centre, Department of NeurologyJawaharlal Institute of Postgraduate Medical Education and ResearchPuducherryIndia
| |
Collapse
|
11
|
Nozohouri S, Sifat AE, Vaidya B, Abbruscato TJ. Novel approaches for the delivery of therapeutics in ischemic stroke. Drug Discov Today 2020; 25:535-551. [PMID: 31978522 DOI: 10.1016/j.drudis.2020.01.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Here, we review novel approaches to deliver neuroprotective drugs to salvageable penumbral brain areas of stroke injury with the goals of offsetting ischemic brain injury and enhancing recovery.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
12
|
Wang W, Liu X, Lu H, Liu L, Wang Y, Yu Y, Zhang T. A method for predicting the success of Pulsinell’s four-vessel occlusion rat model by LDF monitoring of cerebral blood flow decline. J Neurosci Methods 2019; 328:108439. [DOI: 10.1016/j.jneumeth.2019.108439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
|
13
|
Ouk T, Potey C, Maestrini I, Petrault M, Mendyk AM, Leys D, Bordet R, Gautier S. Neutrophils in tPA-induced hemorrhagic transformations: Main culprit, accomplice or innocent bystander? Pharmacol Ther 2019; 194:73-83. [DOI: 10.1016/j.pharmthera.2018.09.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Zhang Z, Zhang L, Ding Y, Han Z, Ji X. Effects of Therapeutic Hypothermia Combined with Other Neuroprotective Strategies on Ischemic Stroke: Review of Evidence. Aging Dis 2018; 9:507-522. [PMID: 29896438 PMCID: PMC5988605 DOI: 10.14336/ad.2017.0628] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 06/28/2017] [Indexed: 12/19/2022] Open
Abstract
Ischemic stroke is a major cause of death and disability globally, and its incidence is increasing. The only treatment approved by the US Food and Drug Administration for acute ischemic stroke is thrombolytic treatment with recombinant tissue plasminogen activator. As an alternative, therapeutic hypothermia has shown excellent potential in preclinical and small clinical studies, but it has largely failed in large clinical studies. This has led clinicians to explore the combination of therapeutic hypothermia with other neuroprotective strategies. This review examines preclinical and clinical progress towards developing highly effective combination therapy involving hypothermia for stroke patients.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Linlei Zhang
- Department of Neurology, the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuchuan Ding
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Zhao Han
- Department of Neurology, the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Granulocyte Colony-Stimulating Factor Alleviates Bacterial-Induced Neuronal Apoptotic Damage in the Neonatal Rat Brain through Epigenetic Histone Modification. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9797146. [PMID: 29484107 PMCID: PMC5816840 DOI: 10.1155/2018/9797146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/07/2017] [Accepted: 11/23/2017] [Indexed: 12/17/2022]
Abstract
Bacterial meningitis during the perinatal period may cause long-term neurological deficits. The study investigated whether bacterial lipopolysaccharide (LPS) derived from E. coli. led to neuronal apoptosis with an impaired performance of long-term cognitive function involving the activation of histone modification in the TNF-α gene promoter. Further, we looked into the therapeutic efficacy of granulocyte colony-stimulating factor (G-CSF) in a neonatal brain suffering from perinatal bacterial meningitis. We applied the following research techniques: neurobehavioral tasks, confocal laser microscopy, chromatin immunoprecipitation, and Western blotting. At postnatal day 10, the animals were subjected to LPS and/or G-CSF. The target brain tissues were then collected at P17. Some animals (P45) were studied using neurobehavioral tasks. The LPS-injected group revealed significantly increased expression of NF-κB phosphorylation and trimethylated H3K4 in the TNFA gene promoter locus. Furthermore, the caspase-3, neuronal apoptosis expression, and an impaired performance in cognitive functions were also found in our study. Such deleterious outcomes described above were markedly alleviated by G-CSF therapy. This study suggests that selective therapeutic action sites of G-CSF through epigenetic regulation in the TNFA gene promoter locus may exert a potentially beneficial role for the neonatal brain suffering from perinatal bacterial-induced meningitis.
Collapse
|
16
|
The Effect of Granulocyte Colony-Stimulating Factor on the Progression of Atherosclerosis in Animal Models: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6705363. [PMID: 29138752 PMCID: PMC5613364 DOI: 10.1155/2017/6705363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 07/12/2017] [Indexed: 11/17/2022]
Abstract
Background Atherosclerosis is a common inflammatory disease. Stem cell and endothelial progenitor cell treatments can improve cardiac function after myocardial infarction. Granulocyte colony-stimulating factor (G-CSF) is a mobilisation agent, mobilising stem cells from the bone marrow to circulation in the blood. G-CSF may constitute a treatment of atherosclerosis. We have conducted meta-analysis to evaluate the current evidence for the effect of G-CSF on the progression of atherosclerosis in animal models and to provide reference for preclinical experiments and future human clinical trials of atherosclerosis treatment. Methods We searched several databases and conducted a meta-analysis across seven articles using a random-effect model. All statistical analyses were performed using Review Manager Version 5.2 and Stata 12.0. Results We found that G-CSF therapy was associated with reduced atherosclerotic lesion area (weighted mean difference (WMD): 7.29%; 95% confidence interval (CI): 2.06-12.52%; P = 0.006). No significant differences in total serum cholesterol (P = 0.54) and triglyceride levels (P = 0.95) were noted in G-CSF treatment groups compared with controls. Multivariable metaregression analysis revealed that the animal type (rabbit, P = 0.022) and frequency of G-CSF administration (>20, P = 0.007) impacted the atherosclerotic lesion area changes. Conclusion The meta-analysis suggested that G-CSF treatment might inhibit the progression of atherosclerosis in animal models.
Collapse
|
17
|
Huang X, Liu Y, Bai S, Peng L, Zhang B, Lu H. Granulocyte colony stimulating factor therapy for stroke: A pairwise meta-analysis of randomized controlled trial. PLoS One 2017; 12:e0175774. [PMID: 28406964 PMCID: PMC5391086 DOI: 10.1371/journal.pone.0175774] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 03/22/2017] [Indexed: 01/27/2023] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) is atherapeutic candidate for stroke that has demonstrated anti-inflammatory and neuroprotective properties. Data from preclinical and clinical studies have suggested the safety and efficacy of G-CSF in stroke; however, the exact effects and utility of this cytokine in patients remain disputed. We performed a meta-analysis of randomized controlled trials of G-CSF in ischemic and hemorrhagic stroke to assess its clinical safety and efficacy. Electronic databases were searched for relevant publications in English and Chinese. A total of 14 trials met the inclusion criteria. G-CSF (cumulative dose range, 1-135μg/kg/day) was tested against placebo in a total of 1037 participants. There was no difference in the rate of mortality between groups (odds ratio, 1.23; 95% confidence interval, 0.76-1.97, p = 0.40). Moreover, the rate of serious adverse events did not differ between groups and provided evidence for the safety of G-CSF administration in stroke patients (odds ratio, 1.11; 95% confidence interval, 0.77-1.61, p = 0.57). No significant outcome benefits were noted with respect to the National Institutes of Health Stroke Scale (mean difference, -0.16; 95% confidence interval, -1.02-0.70, p = 0.72); however, improvements were noted with respect to the Barthel Index (mean difference, 8.65; 95% confidence interval 0.98-16.32; p = 0.03). In conclusion, it appears to be safe in administration of G-CSF, but it will increase leukocyte count. G-CSF was weakly significant benefit with improving the BI scores, while there was no improvement in the NIHSS scores. Larger and more robustly designed trials of G-CSF in stroke are needed to confirm the results.
Collapse
Affiliation(s)
- Xin Huang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Yu Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
- * E-mail:
| | - Shuang Bai
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Lidan Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Boai Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| |
Collapse
|
18
|
Endogenous regeneration: Engineering growth factors for stroke. Neurochem Int 2017; 107:57-65. [PMID: 28411103 DOI: 10.1016/j.neuint.2017.03.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022]
Abstract
Despite the efforts in developing therapeutics for stroke, recombinant tissue plasminogen activator (rtPA) remains the only FDA approved drug for ischemic stroke. Regenerative medicine targeting endogenous growth factors has drawn much interest in the clinical field as it provides potential restoration for the damaged brain tissue without being limited by a narrow therapeutic window. To date, most of the translational studies using regenerative medicines have encountered problems and failures. In this review, we discuss the effects of some trophic factors which include of erythropoietin (EPO), brain derived neurotrophic factor (BDNF), granulocyte-colony stimulating factor (G-CSF), vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), epidermal growth factor (EGF) and heparin binding epidermal growth factor (HB-EGF) in experimental ischemic stroke models and elaborate the lost in translation of the candidate growth factors from bench to bedside. Several new methodologies have been developed to overcome the caveats in translational studies. This review highlights the latest bioengineering approaches including the controlled release and delivery of growth factors by hydrogel-based scaffolds and the enhancement of half-life and selectivity of growth factors by a novel approach facilitated by glycosaminoglycans.
Collapse
|
19
|
Thrombosis, Neuroinflammation, and Poststroke Infection: The Multifaceted Role of Neutrophils in Stroke. J Immunol Res 2017; 2017:5140679. [PMID: 28331857 PMCID: PMC5346374 DOI: 10.1155/2017/5140679] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022] Open
Abstract
Immune cells can significantly predict and affect the clinical outcome of stroke. In particular, the neutrophil-to-lymphocyte ratio was shown to predict hemorrhagic transformation and the clinical outcome of stroke; however, the immunological mechanisms underlying these effects are poorly understood. Neutrophils are the first cells to invade injured tissue following focal brain ischemia. In these conditions, their proinflammatory properties enhance tissue damage and may promote ischemic incidences by inducing thrombus formation. Therefore, they constitute a potential target for therapeutic approaches and prevention of stroke. Indeed, in animal models of focal brain ischemia, neutrophils have been targeted with successful results. However, even in brain lesions, neutrophils also exert beneficial effects, because they are involved in triggering immunological removal of cell debris. Furthermore, intact neutrophil function is essential for maintaining immunological defense against bacterial infections. Several studies have demonstrated that stroke-derived neutrophils displayed impaired bacterial defense capacity. Because infections are known to impair the clinical course of stroke, therapeutic interventions that target neutrophils should preserve or even restore their function outside the central nervous system (CNS). This complex situation requires well-tailored therapeutic approaches that can effectively tackle immune cell invasion in the brain but avoid increasing poststroke infections.
Collapse
|
20
|
Yen JH. Immunomodulatory effect of G-CSF on the CNS infiltrating monocytes in ischemic stroke. Brain Behav Immun 2017; 60:13-14. [PMID: 27765648 DOI: 10.1016/j.bbi.2016.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 10/16/2016] [Indexed: 11/30/2022] Open
Affiliation(s)
- Jui-Hung Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, 2101 E. Coliseum Blvd, Fort Wayne, IN 46805, United States.
| |
Collapse
|
21
|
Weise G, Pösel C, Möller K, Kranz A, Didwischus N, Boltze J, Wagner DC. High-dosage granulocyte colony stimulating factor treatment alters monocyte trafficking to the brain after experimental stroke. Brain Behav Immun 2017; 60:15-26. [PMID: 27524669 DOI: 10.1016/j.bbi.2016.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/26/2016] [Accepted: 08/10/2016] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke elicits a prompt inflammatory response that is characterized by a well-timed recruitment of peripheral immune cells to the brain. Among these, monocytes play a particularly important, but multifaceted role and have been increasingly recognized to affect stroke outcome. Granulocyte colony stimulating factor (GCSF) is known for its immunosuppressive actions on mononuclear cells, but previous studies in the stroke field were mainly confined to its neuroprotective actions. Herein, we investigated whether GCSF affects post-stroke inflammation in a mouse model of focal brain ischemia by modulating monocyte responses. Treatment with GCSF was controlled by vehicle injection, sham surgery and naive animals. Despite a significant monocytosis, high-dosage GCSF reduced the number of brain-infiltrating monocytes/macrophages four days after stroke. Lower numbers of mononuclear phagocytes in the brain were associated with smaller cerebral edema and improved motor outcome after stroke. GCSF treatment over 72h, but not 24h diminished integrin expression on circulating Ly6C+ inflammatory monocytes. In vitro experiments further revealed that GCSF strongly promotes interleukin (IL)-10 secretion by activated mononuclear cells. Blockade of the IL-10 receptor partly reversed GCSF-induced downregulation of integrin surface expression. Overall, our results suggest that high-dosage GCSF mitigates monocyte infiltration after stroke, likely by attenuating integrin-mediated adhesion to the brain endothelium in an IL-10-dependent manner. Lower amounts of mononuclear cells in the brain translate to less severe brain edema and functional impairment and thus support a harmful role of Ly6C+ inflammatory monocytes in the acute stage of stroke.
Collapse
Affiliation(s)
- Gesa Weise
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; University of Leipzig, Department of Neurology, Leipzig, Germany.
| | - Claudia Pösel
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Karoline Möller
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Alexander Kranz
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Nadine Didwischus
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Institute for Biology, Human Biology, University of Leipzig, Leipzig, Germany
| | - Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Fraunhofer Research Institution of Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany; Massachusetts General Hospital and Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA, USA
| | - Daniel-Christoph Wagner
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Institute of Pathology, University Medical Center Mainz, Germany
| |
Collapse
|
22
|
Sandu RE, Balseanu AT, Bogdan C, Slevin M, Petcu E, Popa-Wagner A. Stem cell therapies in preclinical models of stroke. Is the aged brain microenvironment refractory to cell therapy? Exp Gerontol 2017; 94:73-77. [PMID: 28093317 DOI: 10.1016/j.exger.2017.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Stroke is a devastating disease demanding vigorous search for new therapies. Initial enthusiasm to stimulate restorative processes in the ischemic brain by means of cell-based therapies has meanwhile converted into a more balanced view recognizing impediments that may be related to unfavorable age-associated environments. Recent results using a variety of drug, cell therapy or combination thereof suggest that, (i) treatment with Granulocyte-Colony Stimulating Factor (G-CSF) in aged rats has primarily a beneficial effect on functional outcome most likely via supportive cellular processes such as neurogenesis; (ii) the combination therapy, G-CSF with mesenchymal cells (G-CSF+BM-MSC or G-CSF+BM-MNC) did not further improve behavioral indices, neurogenesis or infarct volume as compared to G-CSF alone in aged animals; (iii) better results with regard to integration of transplanted cells in the aged rat environment have been obtained using iPS of human origin; (iv) mesenchymal cells may be used as drug carriers for the aged post-stroke brains. CONCLUSION While the middle aged brain does not seem to impair drug and cell therapies, in a real clinical practice involving older post-stroke patients, successful regenerative therapies would have to be carried out for a much longer time.
Collapse
Affiliation(s)
- Raluca Elena Sandu
- University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania
| | - Adrian Tudor Balseanu
- University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania
| | - Catalin Bogdan
- University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania
| | - Mark Slevin
- Department of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Eugen Petcu
- Griffith University School of Medicine, Gold Coast Campus, QLD 4222, Australia
| | - Aurel Popa-Wagner
- Department of Psychiatry, University Hospital Rostock, Germany; University of Medicine and Pharmacy of Craiova, Chair of Biochemistry, Neurobiology of Aging Group, Romania.
| |
Collapse
|
23
|
Sprigg N, O’Connor R, Woodhouse L, Krishnan K, England TJ, Connell LA, Walker MF, Bath PM. Granulocyte Colony Stimulating Factor and Physiotherapy after Stroke: Results of a Feasibility Randomised Controlled Trial: Stem Cell Trial of Recovery EnhanceMent after Stroke-3 (STEMS-3 ISRCTN16714730). PLoS One 2016; 11:e0161359. [PMID: 27610616 PMCID: PMC5017715 DOI: 10.1371/journal.pone.0161359] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 08/04/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Granulocyte-colony stimulating factor (G-CSF) mobilises endogenous haematopoietic stem cells and enhances recovery in experimental stroke. Recovery may also be dependent on an enriched environment and physical activity. G-CSF may have the potential to enhance recovery when used in combination with physiotherapy, in patients with disability late after stroke. METHODS A pilot 2 x 2 factorial randomised (1:1) placebo-controlled trial of G-CSF (double-blind), and/or a 6 week course of physiotherapy, in 60 participants with disability (mRS >1), at least 3 months after stroke. Primary outcome was feasibility, acceptability and tolerability. Secondary outcomes included death, dependency, motor function and quality of life measured 90 and 365 days after enrolment. RESULTS Recruitment to the trial was feasible and acceptable; of 118 screened patients, 92 were eligible and 32 declined to participate. 60 patients were recruited between November 2011 and July 2013. All participants received some allocated treatment. Although 29 out of 30 participants received all 5 G-CSF/placebo injections, only 7 of 30 participants received all 18 therapy sessions. G-CSF was well tolerated but associated with a tendency to more adverse events than placebo (16 vs 10 patients, p = 0.12) and serious adverse events (SAE) (9 vs 3, p = 0.10). On average, patients received 14 (out of 18 planned) therapy sessions, interquartile range [12, 17]. Only a minority (23%) of participants completed all physiotherapy sessions, a large proportion of sessions (114 of 540, 21%) were cancelled due to patient (94, 17%) and therapist factors (20, 4%). No significant differences in functional outcomes were detected in either the G-CSF or physiotherapy group at day 90 or 365. CONCLUSIONS Delivery of G-CSF is feasible in chronic stroke. However, the study failed to demonstrate feasibility for delivering additional physiotherapy sessions late after stroke therefore a definitive study using this trial design is not supported. Future work should occur earlier after stroke, alongside on-going clinical rehabilitation. TRIAL REGISTRATION ISRCTN.com ISRCTN16714730.
Collapse
Affiliation(s)
- Nikola Sprigg
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Rebecca O’Connor
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Lisa Woodhouse
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Kailash Krishnan
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Timothy J. England
- Division of Medical Sciences and GEM, University of Nottingham, Nottingham, United Kingdom
| | - Louise A. Connell
- School of Health, University of Central Lancashire, Preston, United Kingdom
| | - Marion F. Walker
- Division of Rehabilitation and Ageing, University of Nottingham, Nottingham, United Kingdom
| | - Philip M. Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
24
|
Strecker JK, Olk J, Hoppen M, Gess B, Diederich K, Schmidt A, Schäbitz WR, Schilling M, Minnerup J. Combining Growth Factor and Bone Marrow Cell Therapy Induces Bleeding and Alters Immune Response After Stroke in Mice. Stroke 2016; 47:852-62. [PMID: 26839353 DOI: 10.1161/strokeaha.115.011230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/31/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Bone marrow cell (BMC)-based therapies, either the transplantation of exogenous cells or stimulation of endogenous cells by growth factors like the granulocyte colony-stimulating factor (G-CSF), are considered a promising means of treating stroke. In contrast to large preclinical evidence, however, a recent clinical stroke trial on G-CSF was neutral. We, therefore, aimed to investigate possible synergistic effects of co-administration of G-CSF and BMCs after experimental stroke in mice to enhance the efficacy compared with single treatments. METHODS We used an animal model for experimental stroke as paradigm to study possible synergistic effects of co-administration of G-CSF and BMCs on the functional outcome and the pathophysiological mechanism. RESULTS G-CSF treatment alone led to an improved functional outcome, a reduced infarct volume, increased blood vessel stabilization, and decreased overall inflammation. Surprisingly, the combination of G-CSF and BMCs abrogated G-CSFs' beneficial effects and resulted in increased hemorrhagic infarct transformation, altered blood-brain barrier, excessive astrogliosis, and altered immune cell polarization. These increased rates of infarct bleeding were mainly mediated by elevated matrix metalloproteinase-9-mediated blood-brain barrier breakdown in G-CSF- and BMCs-treated animals combined with an increased number of dilated and thus likely more fragile vessels in the subacute phase after cerebral ischemia. CONCLUSIONS Our results provide new insights into both BMC-based therapies and immune cell biology and help to understand potential adverse and unexpected side effects.
Collapse
Affiliation(s)
- Jan-Kolja Strecker
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.).
| | - Joanna Olk
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Maike Hoppen
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Burkhard Gess
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Kai Diederich
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Antje Schmidt
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Wolf-Rüdiger Schäbitz
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Matthias Schilling
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| | - Jens Minnerup
- From the Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany (J.-K.S., J.O., M.H., B.G., K.D., A.S., M.S., J.M.); and EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.)
| |
Collapse
|
25
|
Hashemzaei M, Imen Shahidi M, Moallem SA, Abnous K, Ghorbani M, Mohamadpour AH. Modulation of JAK2, STAT3 and Akt1 proteins by granulocyte colony stimulating factor following carbon monoxide poisoning in male rat. Drug Chem Toxicol 2016; 39:375-9. [DOI: 10.3109/01480545.2015.1123267] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
26
|
Abstract
Over recent decades, experimental and clinical stroke studies have identified a number of neurorestorative treatments that stimulate neural plasticity and promote functional recovery. In contrast to the acute stroke treatments thrombolysis and endovascular thrombectomy, neurorestorative treatments are still effective when initiated days after stroke onset, which makes them applicable to virtually all stroke patients. In this article, selected physical, pharmacological and cell-based neurorestorative therapies are discussed, with special emphasis on interventions that have already been transferred from the laboratory to the clinical setting. We explain molecular and structural processes that promote neural plasticity, discuss potential limitations of neurorestorative treatments, and offer a speculative viewpoint on how neurorestorative treatments will evolve.
Collapse
Affiliation(s)
- Antje Schmidt
- a Department of Neurology , University of Münster , Münster , Germany
| | - Jens Minnerup
- a Department of Neurology , University of Münster , Münster , Germany
| |
Collapse
|
27
|
Neubauer V, Wegleiter K, Posod A, Urbanek M, Wechselberger K, Kiechl-Kohlendorfer U, Keller M, Griesmaier E. Delayed application of the haematopoietic growth factors G-CSF/SCF and FL reduces neonatal excitotoxic brain injury. Brain Res 2016; 1634:94-103. [PMID: 26772988 DOI: 10.1016/j.brainres.2015.12.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/03/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Developmental brain injury results in cognitive and motor deficits in the preterm infant. Enhanced glutamate release and subsequent receptor activation are major pathogenetic factors. The effect of haematopoietic growth factors, such as granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF) and flt-3 ligand (FL) on neonatal brain injury is controversially discussed. Timing of treatment is known to be a crucial factor. Based on the hypothesis that an exacerbation of injury is caused by administration of substances in the acute phase, the objective of this study was to evaluate the effect of delayed administration of G-CSF/SCF and FL to protect against excitotoxic brain injury in vivo. METHODS In an established neonatal mouse model of excitotoxic brain injury, we evaluated the effect of daily intraperitoneal doses of G-CSF/SCF or FL, starting 60 h after the excitotoxic insult. RESULTS Intraperitoneal injections of G-CSF/SCF and FL, given 60 h after the excitotoxic insult, significantly reduced lesion size at postnatal days 10, 18 and 90. G-CSF/SCF treatment resulted in a decrease in apoptotic cell death indicated by reduced caspase-3 activation. G-CSF/SCF and FL treatment did not affect apoptosis-inducing factor-dependent apoptosis or cell proliferation. CONCLUSION We show that delayed systemic treatment with the haematopoietic growth factors G-CSF/SCF and FL protects against N-methyl-D-aspartate receptor-mediated developmental excitotoxic brain damage. Our results suggest that neuroprotective effects in this neonatal animal model of excitotoxic brain injury depend on the timing of drug administration after the insult.
Collapse
Affiliation(s)
- Vera Neubauer
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Karina Wegleiter
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Anna Posod
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Martina Urbanek
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Karina Wechselberger
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Ursula Kiechl-Kohlendorfer
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Matthias Keller
- Kinderklinik Dritter Orden, Munich Technical University, Bischof Altmann-Strasse 9, 94032 Passau, Germany
| | - Elke Griesmaier
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria.
| |
Collapse
|
28
|
Minnerup J, Zentsch V, Schmidt A, Fisher M, Schäbitz WR. Methodological Quality of Experimental Stroke Studies Published in the
Stroke
Journal. Stroke 2016; 47:267-72. [DOI: 10.1161/strokeaha.115.011695] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/04/2015] [Indexed: 02/01/2023]
Affiliation(s)
- Jens Minnerup
- From the Department of Neurology, University of Münster, Münster, Germany (J.M., V.Z., A.S.); Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.); and Department of Neurology, Bethel-Evangelisches Krankenhaus, Bielefeld, Germany (W.-R.S)
| | - Verena Zentsch
- From the Department of Neurology, University of Münster, Münster, Germany (J.M., V.Z., A.S.); Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.); and Department of Neurology, Bethel-Evangelisches Krankenhaus, Bielefeld, Germany (W.-R.S)
| | - Antje Schmidt
- From the Department of Neurology, University of Münster, Münster, Germany (J.M., V.Z., A.S.); Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.); and Department of Neurology, Bethel-Evangelisches Krankenhaus, Bielefeld, Germany (W.-R.S)
| | - Marc Fisher
- From the Department of Neurology, University of Münster, Münster, Germany (J.M., V.Z., A.S.); Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.); and Department of Neurology, Bethel-Evangelisches Krankenhaus, Bielefeld, Germany (W.-R.S)
| | - Wolf-Rüdiger Schäbitz
- From the Department of Neurology, University of Münster, Münster, Germany (J.M., V.Z., A.S.); Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.); and Department of Neurology, Bethel-Evangelisches Krankenhaus, Bielefeld, Germany (W.-R.S)
| |
Collapse
|
29
|
Kleikers PWM, Hooijmans C, Göb E, Langhauser F, Rewell SSJ, Radermacher K, Ritskes-Hoitinga M, Howells DW, Kleinschnitz C, HHW Schmidt H. A combined pre-clinical meta-analysis and randomized confirmatory trial approach to improve data validity for therapeutic target validation. Sci Rep 2015; 5:13428. [PMID: 26310318 PMCID: PMC4550831 DOI: 10.1038/srep13428] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/27/2015] [Indexed: 12/30/2022] Open
Abstract
Biomedical research suffers from a dramatically poor translational success. For example, in ischemic stroke, a condition with a high medical need, over a thousand experimental drug targets were unsuccessful. Here, we adopt methods from clinical research for a late-stage pre-clinical meta-analysis (MA) and randomized confirmatory trial (pRCT) approach. A profound body of literature suggests NOX2 to be a major therapeutic target in stroke. Systematic review and MA of all available NOX2(-/y) studies revealed a positive publication bias and lack of statistical power to detect a relevant reduction in infarct size. A fully powered multi-center pRCT rejects NOX2 as a target to improve neurofunctional outcomes or achieve a translationally relevant infarct size reduction. Thus stringent statistical thresholds, reporting negative data and a MA-pRCT approach can ensure biomedical data validity and overcome risks of bias.
Collapse
Affiliation(s)
- Pamela WM. Kleikers
- Department of Pharmacology, CARIM, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Carlijn Hooijmans
- SYRCLE at Central Animal Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Eva Göb
- Neurologische Klinik und Poliklinik der Universitätsklinik Würzburg, Würzburg, Germany
| | - Friederike Langhauser
- Neurologische Klinik und Poliklinik der Universitätsklinik Würzburg, Würzburg, Germany
| | - Sarah SJ. Rewell
- Florey Institute of Neuroscience and Mental Health, Austin Health, Melbourne, Victoria, Australia
| | - Kim Radermacher
- Department of Pharmacology, CARIM, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Merel Ritskes-Hoitinga
- SYRCLE at Central Animal Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - David W. Howells
- Florey Institute of Neuroscience and Mental Health, Austin Health, Melbourne, Victoria, Australia
| | | | - Harald HHW Schmidt
- Department of Pharmacology, CARIM, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| |
Collapse
|
30
|
The Efficacy and Safety of Granulocyte Colony-Stimulating Factor for Patients with Stroke. J Stroke Cerebrovasc Dis 2015; 24:1701-8. [DOI: 10.1016/j.jstrokecerebrovasdis.2014.11.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 11/18/2022] Open
|
31
|
Riecke J, Johns KM, Cai C, Vahidy FS, Parsha K, Furr-Stimming E, Schiess M, Savitz SI. A Meta-Analysis of Mesenchymal Stem Cells in Animal Models of Parkinson's Disease. Stem Cells Dev 2015; 24:2082-90. [PMID: 26134374 DOI: 10.1089/scd.2015.0127] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multiple studies have been performed to evaluate the effects of mesenchymal stem cells (MSCs) in animal models of Parkinson's disease (PD). We performed a meta-analysis to estimate the treatment effect of unmodified MSCs on behavioral outcomes in preclinical studies of PD. We performed a systematic literature search to identify studies that used behavioral testing to evaluate the treatment effect of unmodified MSCs in PD models. Meta-analysis was used to determine pooled effect size for rotational behavior and limb function, and meta-regression was performed to explore sources of heterogeneity. Twenty-five studies, including three delivery routes, a wide range of doses, and multiple PD models, were examined. Significant improvement was seen in the pooled standardized mean difference (SMD) for both rotational behavior [SMD: 1.24, 95% confidence interval (95% CI): 0.84, 1.64] and limb function (SMD: 0.84, 95% CI: 0.01, 1.66). Using meta-regression, intravenous administration and higher dose had a larger effect on limb function. Treatment with MSCs improves behavioral outcomes in PD models. Our analyses suggest that MSCs could be considered for early-stage clinical trials in the treatment of PD.
Collapse
Affiliation(s)
- Jenny Riecke
- 1 Department of Neurology, University of Texas-Houston Medical School , Houston, Texas
| | - Katherine M Johns
- 1 Department of Neurology, University of Texas-Houston Medical School , Houston, Texas
| | - Chunyan Cai
- 2 Division of Clinical and Translational Sciences, Department of Internal Medicine, University of Texas-Houston Medical School , Houston, Texas
| | | | - Kaushik Parsha
- 1 Department of Neurology, University of Texas-Houston Medical School , Houston, Texas
| | - Erin Furr-Stimming
- 1 Department of Neurology, University of Texas-Houston Medical School , Houston, Texas
| | - Mya Schiess
- 1 Department of Neurology, University of Texas-Houston Medical School , Houston, Texas
| | - Sean I Savitz
- 1 Department of Neurology, University of Texas-Houston Medical School , Houston, Texas
| |
Collapse
|
32
|
Ghahari L, Safari M, Joghataei MT, Mehdizadeh M, Soleimani M. Effect of combination therapy using hypothermia and granulocyte colony-stimulating factor in a rat transient middle cerebral artery occlusion model. IRANIAN BIOMEDICAL JOURNAL 2015; 18:239-44. [PMID: 25326023 PMCID: PMC4225064 DOI: 10.6091/ibj.13852.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Stroke is the third leading cause of death. Hypothermia has been recognized as an effective method in reducing brain injury. In this study, we assessed the effects of granulocyte colony-stimulating factor (G-CSF) as a neuroprotective agent and mild hypothermia on mortality, behavioral function, infarct volume, and brain edema in Wistar rats. Methods: Forty male rats were used in five groups (eight rats in each group): control, hypothermy, G-CSF, combination hypothermy + CSF, and sham. Rats were anesthetized by injection of chloral hydrate (400 mg/kg) intraperitoneally. Transient cerebral ischemia was induced by 60-min intraluminal occlusion of left middle cerebral artery. Hypothermia, initiated at the time of reperfusion and G-CSF was started one hour after reperfusion at a dose of 15 mg/kg subcutaneously. The motor behavior was measured using Garcia’s index and animals were assigned for the assessments of infarction, brain swelling, and mortality rate. Results: The mortality was 38.46% (control group) and reduced in other groups. Neurological deficit score of control group (40.31 ± 1.56) was significantly lower than in treatment groups. The total cerebral infarct volume of treatment group was significantly lower than control group (43.96 ± 44.05 mm3). Treatment with hypothermy plus G-CSF (2.69 ± 0.24%) could significantly reduce brain swelling volume than other treatment groups. Conclusion: Our major finding is that mild hypothermic treatment plus G-CSF significantly reduced mortality rate and edema and improved neurological function. The results suggest that the combination of hypothermia and G-CSF is more effectively than other treatment groups being used alone.
Collapse
Affiliation(s)
- Laya Ghahari
- Dept. of Anatomy, Medical School, Iran University of Medical Science, Tehran, Iran.,Dept. of Anatomy, Medical School, AJA University of Medical Sciences, Tehran, Iran
| | - Manouchehr Safari
- Dept. of Anatomy, Medical School, Semnan
University of Medical Science, Semnan, Iran
| | - Mohamad Taghi Joghataei
- Cellular and Molecular Research Center, Faculty of Advanced Technology in Medicine, Dept. of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Cellular and Molecular Research Center, Faculty of Advanced Technology in Medicine, Dept. of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Dept. of Anatomy, Medical School, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
33
|
Neuroprotection for ischaemic stroke: Current status and challenges. Pharmacol Ther 2015; 146:23-34. [DOI: 10.1016/j.pharmthera.2014.09.003] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/31/2022]
|
34
|
Beom J, Kim W, Han TR, Seo KS, Oh BM. Concurrent use of granulocyte-colony stimulating factor with repetitive transcranial magnetic stimulation did not enhance recovery of function in the early subacute stroke in rats. Neurol Sci 2014; 36:771-7. [PMID: 25528461 DOI: 10.1007/s10072-014-2046-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 12/15/2014] [Indexed: 11/28/2022]
Abstract
We investigated the additive effect of repetitive transcranial magnetic stimulation (rTMS) combined with granulocyte-colony stimulating factor (G-CSF) on functional outcome in the early subacute phase of stroke. Seven-week-old male rats were subjected to permanent middle cerebral artery occlusion (MCAo) and were divided into four groups: normal saline administration with sham rTMS (group 1, n = 15), G-CSF administration with sham rTMS (group 2, n = 15), G-CSF with 1 Hz rTMS (group 3, n = 14), and G-CSF with 20 Hz rTMS (group 4, n = 15). Animals received G-CSF or saline for 5 days from the day of MCAo and were concurrently treated with 20-min rTMS on their lesioned hemisphere for 2 weeks. Neurological functional score was worse in group 4 compared to that in group 2 on day 15. In Western blots conducted on day 25, phosphorylation of endothelial nitric oxide synthase was markedly lower in groups 2, 3, and 4 than that in group 1 in the ischemic border zone. PECAM-1 expression at ischemic core was lower in groups 4 than in group 2. Caspase-3 expression was markedly higher in groups 4 than in group 1, 2, 3 at ischemic core. Iba1 expression was higher in groups 4 than in group 1, 2 at ischemic core. G-CSF combined with rTMS administered in the early subacute phase of ischemic stroke may exert a hazardous effect on functional recovery, possibly due to impaired angiogenic mechanism, decreased cell survival, and increased inflammation.
Collapse
Affiliation(s)
- Jaewon Beom
- Department of Rehabilitation Medicine, Chungnam National University Hospital, Daejeon, 301-721, Republic of Korea
| | | | | | | | | |
Collapse
|
35
|
Diederich K, Schmidt A, Beuker C, Strecker JK, Wagner DC, Boltze J, Schäbitz WR, Minnerup J. Granulocyte colony-stimulating factor (G-CSF) treatment in combination with transplantation of bone marrow cells is not superior to G-CSF treatment alone after cortical stroke in spontaneously hypertensive rats. Front Cell Neurosci 2014; 8:411. [PMID: 25538562 PMCID: PMC4255603 DOI: 10.3389/fncel.2014.00411] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/12/2014] [Indexed: 12/02/2022] Open
Abstract
Granulocyte-colony stimulating factor (G-CSF) and bone marrow derived mononuclear cells (BM-MNCs) have both been shown to improve functional outcome following experimental stroke. These effects are associated with increased angiogenesis and neurogenesis. In the present study, we aimed to determine synergistic effects of G-CSF and BM-NMC treatment on long-term structural and functional recovery after photothrombotic stroke. To model the etiology of stroke more closely, we used spontaneously hypertensive (SH) rats in our experiment. Bone marrow derived mononuclear cells transplantation was initiated 1 h after the onset of photothrombotic stroke. Repeated G-CSF treatment commenced immediately after BM-MNC treatment followed by daily injections for five consecutive days. The primary endpoint was functional outcome after ischemia. Secondary endpoints included analysis of neurogenesis and angiogenesis as well as determination of infarct size. Granulocyte-colony stimulating factor treated rats, either in combination with BM-MNC or alone showed improved somatosensory but not gross motor function following ischemia. No beneficial effect of BM-MNC monotherapy was found. Infarct volumes were comparable in all groups. In contrast to previous studies, which used healthy animals, post-stroke neurogenesis and angiogenesis were not enhanced by G-CSF. In conclusion, the combination of G-CSF and BM-MNC was not more effective than G-CSF alone. The reduced efficacy of G-CSF treatment and the absence of any beneficial effect of BM-MNC transplantation might be attributed to hypertension-related morbidity.
Collapse
Affiliation(s)
- Kai Diederich
- Department of Neurology, University of Münster Münster, Germany
| | - Antje Schmidt
- Department of Neurology, University of Münster Münster, Germany
| | - Carolin Beuker
- Department of Neurology, University of Münster Münster, Germany
| | | | - Daniel-Christoph Wagner
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany ; Translational Center for Regenerative Medicine, University of Leipzig Leipzig, Germany
| | - Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany ; Translational Center for Regenerative Medicine, University of Leipzig Leipzig, Germany
| | | | - Jens Minnerup
- Department of Neurology, University of Münster Münster, Germany
| |
Collapse
|
36
|
Sun BL, He MQ, Han XY, Sun JY, Yang MF, Yuan H, Fan CD, Zhang S, Mao LL, Li DW, Zhang ZY, Zheng CB, Yang XY, Li YV, Stetler RA, Chen J, Zhang F. Intranasal Delivery of Granulocyte Colony-Stimulating Factor Enhances Its Neuroprotective Effects Against Ischemic Brain Injury in Rats. Mol Neurobiol 2014; 53:320-330. [DOI: 10.1007/s12035-014-8984-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/03/2014] [Indexed: 12/20/2022]
|
37
|
Pösel C, Scheibe J, Kranz A, Bothe V, Quente E, Fröhlich W, Lange F, Schäbitz WR, Minnerup J, Boltze J, Wagner DC. Bone marrow cell transplantation time-dependently abolishes efficacy of granulocyte colony-stimulating factor after stroke in hypertensive rats. Stroke 2014; 45:2431-7. [PMID: 24984745 DOI: 10.1161/strokeaha.113.004460] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE We aimed to determine a possible synergistic effect of granulocyte colony-stimulating factor (G-CSF) and bone marrow-derived mononuclear cells (BM MNC) after stroke in spontaneously hypertensive rats. METHODS Male spontaneously hypertensive rats were subjected to middle cerebral artery occlusion and randomly assigned to daily injection of 50 μg/kg G-CSF for 5 days starting 1 hour after stroke (groups 1, 2, and 3) with additional intravenous transplantation of 1.5×10E7 BM MNC per kilogram at 6 hours (group 2) or 48 hours (group 3) after stroke, or control treatment (group 4). Circulating leukocyte counts and functional deficits, infarct volume, and brain edema were repeatedly assessed in the first week and first month. RESULTS G-CSF treatment led to a significant neutrophilia, to a reversal of postischemic depression of circulating leukocytes, and to a significantly improved functional recovery without affecting the infarct volume or brain edema. BM MNC cotransplantation was neutral after 6 hours, but reversed the functional effect of G-CSF after 48 hours. Short-term investigation of combined G-CSF and BM MNC treatment at 48 hours indicated splenic accumulation of granulocytes and transplanted cells, accompanied by a significant rise of granulocytes in the circulation and the ischemic brain. CONCLUSIONS G-CSF improved functional recovery in spontaneously hypertensive rats, but this effect was abolished by cotransplantation of BM MNC after 48 hours. In the spleen, transplanted cells may hinder the clearance of granulocytes that were massively increased by G-CSF. Increased circulation and infiltration of granulocytes into the ischemic brain may be detrimental for stroke outcome.
Collapse
Affiliation(s)
- Claudia Pösel
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.).
| | - Johanna Scheibe
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Alexander Kranz
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Viktoria Bothe
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Elfi Quente
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Wenke Fröhlich
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Franziska Lange
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Wolf-Rüdiger Schäbitz
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Jens Minnerup
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Johannes Boltze
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| | - Daniel-Christoph Wagner
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (C.P., J.S., A.K., E.Q., W.F., F.L., J.B., D.-C.W.); Translational Centre for Regenerative Medicine, Leipzig, Germany (A.K., V.B., E.Q., W.F., J.B., D.-C.W.); EVK Bielefeld, Bethel, Neurologische Klinik, Bielefeld, Germany (W.-R.S.); Department of Neurology, University of Münster, Germany (J.M.); and Massachusetts General Hospital and Harvard Medical School, Boston (J.B.)
| |
Collapse
|
38
|
Gautier S, Ouk T, Tagzirt M, Lefebvre C, Laprais M, Pétrault O, Dupont A, Leys D, Bordet R. Impact of the neutrophil response to granulocyte colony-stimulating factor on the risk of hemorrhage when used in combination with tissue plasminogen activator during the acute phase of experimental stroke. J Neuroinflammation 2014; 11:96. [PMID: 24885160 PMCID: PMC4059099 DOI: 10.1186/1742-2094-11-96] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/13/2014] [Indexed: 01/06/2023] Open
Abstract
Background Granulocyte colony-stimulating factor (G-CSF) is a pharmacologic agent inducing neutrophil mobilization and a new candidate for neuroprotection and neuroregeneration in stroke. Its effects when used in combination with tissue plasminogen activator (tPA) were explored during the acute phase of ischemic stroke. Methods We used a middle cerebral artery occlusion (MCAO) model of cerebral ischemia, associated with treatment with tPA, in male spontaneously hypertensive rats (SHR). Granulocyte colony-stimulating factor (G-CSF; 60 μg/kg) was injected just before tPA. Neutrophil response in peripheral blood and in the infarct area was quantified in parallel to the infarct volume. Protease matrix metallopeptidase 9 (MMP-9) release from circulating neutrophils was analyzed by immunochemistry and zymography. Vascular reactivity and hemorrhagic volume in the infarct area was also assessed. Results Twenty four hours after ischemia and tPA, G-CSF administration induced a significant increase of neutrophils in peripheral blood (P <0.05). At 72 hours post-ischemia, G-CSF was significantly associated with an increased risk of hemorrhage in the infarct area (2.5 times more likely; P <0.05) and significant cerebral endothelium-dependent dysfunction. Ex vivo, an increased MMP-9 release from neutrophils after tPA administration correlated to the increased hemorrhagic risk (P <0.05). In parallel, G-CSF administration was associated with a decreased neutrophil infiltration in the infarct area (-50%; P <0.05), with a concomitant significant neuroprotective effect (infarct volume: -40%; P <0.05). Conclusions We demonstrate that G-CSF potentiates the risk of hemorrhage in experimental stroke when used in combination with tPA by inducing neutrophilia. This effect is concomitant to an increased MMP-9 release from peripheral neutrophils induced by the tPA treatment. These results highlight the potential hemorrhagic risk of associating G-CSF to thrombolysis during the acute phase of stroke.
Collapse
Affiliation(s)
- Sophie Gautier
- EA 1046 - Département de Pharmacologie médicale, Université de Lille 2 - Faculté de Médecine, 1 place de Verdun, Lille cedex F-59037, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wagner DC, Pösel C, Schulz I, Schicht G, Boltze J, Lange F, Scheibe J, Möller K, Weise G. Allometric dose retranslation unveiled substantial immunological side effects of granulocyte colony-stimulating factor after stroke. Stroke 2014; 45:623-6. [PMID: 24407949 DOI: 10.1161/strokeaha.113.003812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND PURPOSE Granulocyte colony-stimulating factor (GCSF) showed robust neuroprotective and neuroregenerative properties after stroke in rodents but failed to meet study end points in patients. Because immunologic side effects of GCSF may have escaped preclinical testing because of nonallometric dose translation, we hypothesized those as possible reasons. METHODS Stroke was induced in C57BL/6 mice by 45-minute filament middle cerebral artery occlusion. GCSF was administered at 50 and 832.5 μg/kg body weight. Treatment was controlled by vehicle injection, sham surgery, and naive animals. Immune cell counts were assessed in blood, spleen, and brain by multidimensional flow cytometry 1 day after stroke. RESULTS High-dose GCSF significantly altered myeloid and T-cell subpopulations in blood and spleen and caused a tremendous increase of monocytes/macrophages infiltrating the ischemic brain. CONCLUSIONS Dose-dependent immunomodulation superimposes central nervous system-specific effects of GCSF after stroke. Adaption of dose or treatment time may overcome this drawback.
Collapse
Affiliation(s)
- Daniel-Christoph Wagner
- From the Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany (D.-C.W., C.P., I.S., G.S., J.B., F.L., J.S., K.M., G.W.); Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany (D.-C.W., J.B., G.W.); and Stroke and Neurovascular Regulation Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, MA (J.B.)
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ridwan S, Bauer H, Frauenknecht K, Hefti K, von Pein H, Sommer CJ. Distribution of the hematopoietic growth factor G-CSF and its receptor in the adult human brain with specific reference to Alzheimer's disease. J Anat 2014; 224:377-91. [PMID: 24387791 DOI: 10.1111/joa.12154] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2013] [Indexed: 11/29/2022] Open
Abstract
The granulocyte colony-stimulating factor (G-CSF), being a member of the hematopoietic growth factor family, is also critically involved in controlling proliferation and differentiation of neural stem cells. Treatment with G-CSF has been shown to result in substantial neuroprotective and neuroregenerative effects in various experimental models of acute and chronic diseases of the central nervous system. Although G-CSF has been tested in a clinical study for treatment of acute ischemic stroke, there is only fragmentary data on the distribution of this cytokine and its receptor in the human brain. Therefore, the present study was focused on the immunohistochemical analysis of the protein expression of G-CSF and its receptor (G-CSF R) in the adult human brain. Since G-CSF has been shown not only to exert neuroprotective effects in animal models of Alzheimer's disease (AD) but also to be a candidate for clinical treatment, we have also placed an emphasis on the regulation of these molecules in this neurodegenerative disease. One major finding is that both G-CSF and G-CSF R were ubiquitously but not uniformly expressed in neurons throughout the CNS. Protein expression of G-CSF and G-CSF R was not restricted to neurons but was also detectable in astrocytes, ependymal cells, and choroid plexus cells. However, the distribution of G-CSF and G-CSF R did not substantially differ between AD brains and control, even in the hippocampus, where early neurodegenerative changes typically occur.
Collapse
Affiliation(s)
- Sami Ridwan
- Department of Neuropathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Taguchi A, Kasahara Y, Matsuyama T. Letter by Taguchi et al Regarding Article, “Granulocyte Colony-Stimulating Factor in Patients With Acute Ischemic Stroke: Results of the AX200 for Ischemic Stroke Trial”. Stroke 2014; 45:e8. [DOI: 10.1161/strokeaha.113.003683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Hyogo, Japan
| | - Yukiko Kasahara
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Hyogo, Japan
| | - Tomohiro Matsuyama
- Laboratory of Neurogenesis and CNS Repair, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Hyogo, Japan
| |
Collapse
|
42
|
Schmidt A, Wellmann J, Schilling M, Strecker JK, Sommer C, Schäbitz WR, Diederich K, Minnerup J. Meta-analysis of the Efficacy of Different Training Strategies in Animal Models of Ischemic Stroke. Stroke 2014; 45:239-47. [DOI: 10.1161/strokeaha.113.002048] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Antje Schmidt
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Jürgen Wellmann
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Matthias Schilling
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Jan-Kolja Strecker
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Clemens Sommer
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Wolf-Rüdiger Schäbitz
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Kai Diederich
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| | - Jens Minnerup
- From the Department of Neurology (A.S., M.S., J.-K.S., K.D., J.M.) and Institute of Epidemiology and Social Medicine (J.W.), University of Münster, Münster, Germany; Department of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany (C.S.); and Department of Neurology, Bethel-EvKB, Bielefeld, Germany (W.-R.S.)
| |
Collapse
|
43
|
Neurogenesis recovery induced by granulocyte-colony stimulating factor in neonatal rat brain after perinatal hypoxia. Pediatr Neonatol 2013; 54:380-8. [PMID: 23791015 DOI: 10.1016/j.pedneo.2013.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 03/14/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Perinatal hypoxia can lead to a wide range of neurological deficits depending on the differential vulnerability of the involved brain regions to oxygen deprivation. It remains unclear whether the differential vulnerability to oxygen deprivation leads to altered neurogenesis in the neonatal brain after perinatal hypoxia. The primary objective was to investigate whether perinatal hypoxia induces deleterious changes in neurogenesis within three representative brain regions (dentate gyrus of the hippocampus, midbrain, and temporal cortex), with regards to common pathological areas clinically. The secondary objective was to investigate whether granulocyte-colony stimulating factor (G-CSF) therapy exerts beneficial effects in neurogenesis in neonatal rat brains subjected to experimental perinatal hypoxia. MATERIALS AND METHODS Rat pups were subjected to experimental perinatal hypoxia on the tenth day of life (P10). They were then given G-CSF (30 μg/kg, single injection/day, intraperitoneal injection, P11-16). The neurogenesis efficacy was analyzed on P17 and the radial-arm maze task, a memory task for higher cognitive functions such as problem-solving abilities, was evaluated on P37-58. RESULTS Perinatal hypoxia caused a significant decrease in neurogenesis within the three representative brain regions, and this deleterious outcome was alleviated by G-CSF (p < 0.05). In addition, the G-CSF therapy markedly improved the decreased performance of long-term cognitive functions induced by perinatal hypoxia (p < 0.05). CONCLUSION This study suggests that G-CSF may be a potentially beneficial therapy, at least in part, through universal recovery of neurogenesis effects in the neonatal brain after perinatal hypoxia insult.
Collapse
|
44
|
Jellema RK, Lima Passos V, Ophelders DRMG, Wolfs TGAM, Zwanenburg A, De Munter S, Nikiforou M, Collins JJP, Kuypers E, Bos GMJ, Steinbusch HW, Vanderlocht J, Andriessen P, Germeraad WTV, Kramer BW. Systemic G-CSF attenuates cerebral inflammation and hypomyelination but does not reduce seizure burden in preterm sheep exposed to global hypoxia-ischemia. Exp Neurol 2013; 250:293-303. [PMID: 24120465 DOI: 10.1016/j.expneurol.2013.09.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 09/24/2013] [Accepted: 09/27/2013] [Indexed: 01/26/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is common in preterm infants, but currently no curative therapy is available. Cell-based therapy has a great potential in the treatment of hypoxic-ischemic preterm brain injury. Granulocyte-colony stimulating factor (G-CSF) is known to mobilize endogenous hematopoietic stem cells (HSC) and promotes proliferation of endogenous neural stem cells. On these grounds, we hypothesized that systemic G-CSF would be neuroprotective in a large translational animal model of hypoxic-ischemic injury in the preterm brain. Global hypoxia-ischemia (HI) was induced by transient umbilical cord occlusion in instrumented preterm sheep. G-CSF treatment (100μg/kg intravenously, during five consecutive days) was started one day before the global HI insult to ascertain mobilization of endogenous stem cells within the acute phase after global HI. Mobilization of HSC and neutrophils was studied by flow cytometry. Brain sections were stained for microglia (IBA-1), myelin basic protein (MBP) and myeloperoxidase (MPO) to study microglial proliferation, white matter injury and neutrophil invasion respectively. Electrographic seizure activity was analyzed using amplitude-integrated electroencephalogram (aEEG). G-CSF effectively mobilized CD34-positive HSC in the preterm sheep. In addition, G-CSF caused marked mobilization of neutrophils, but did not influence enhanced invasion of neutrophils into the preterm brain after global HI. Microglial proliferation and hypomyelination following global HI were reduced as a result of G-CSF treatment. G-CSF did not cause a reduction of the electrographic seizure activity after global HI. In conclusion, G-CSF induced mobilization of endogenous stem cells which was associated with modulation of the cerebral inflammatory response and reduced white matter injury in an ovine model of preterm brain injury after global HI. G-CSF treatment did not improve neuronal function as shown by seizure analysis. Our study shows that G-CSF treatment has neuroprotective potential following hypoxic-ischemic injury in the preterm brain.
Collapse
Affiliation(s)
- Reint K Jellema
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands; Department of Pediatrics, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Krauth D, Woodruff TJ, Bero L. Instruments for assessing risk of bias and other methodological criteria of published animal studies: a systematic review. ENVIRONMENTAL HEALTH PERSPECTIVES 2013; 121:985-92. [PMID: 23771496 PMCID: PMC3764080 DOI: 10.1289/ehp.1206389] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 06/10/2013] [Indexed: 05/07/2023]
Abstract
BACKGROUND Results from animal toxicology studies are critical to evaluating the potential harm from exposure to environmental chemicals or the safety of drugs prior to human testing. However, there is significant debate about how to evaluate the methodology and potential biases of the animal studies. There is no agreed-upon approach, and a systematic evaluation of current best practices is lacking. OBJECTIVE We performed a systematic review to identify and evaluate instruments for assessing the risk of bias and/or other methodological criteria of animal studies. METHOD We searched Medline (January 1966-November 2011) to identify all relevant articles. We extracted data on risk of bias criteria (e.g., randomization, blinding, allocation concealment) and other study design features included in each assessment instrument. DISCUSSION Thirty distinct instruments were identified, with the total number of assessed risk of bias, methodological, and/or reporting criteria ranging from 2 to 25. The most common criteria assessed were randomization (25/30, 83%), investigator blinding (23/30, 77%), and sample size calculation (18/30, 60%). In general, authors failed to empirically justify why these or other criteria were included. Nearly all (28/30, 93%) of the instruments have not been rigorously tested for validity or reliability. CONCLUSION Our review highlights a number of risk of bias assessment criteria that have been empirically tested for animal research, including randomization, concealment of allocation, blinding, and accounting for all animals. In addition, there is a need for empirically testing additional methodological criteria and assessing the validity and reliability of a standard risk of bias assessment instrument.
Collapse
Affiliation(s)
- David Krauth
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA 94118, USA
| | | | | |
Collapse
|
46
|
Ringelstein EB, Thijs V, Norrving B, Chamorro A, Aichner F, Grond M, Saver J, Laage R, Schneider A, Rathgeb F, Vogt G, Charissé G, Fiebach JB, Schwab S, Schäbitz WR, Kollmar R, Fisher M, Brozman M, Skoloudik D, Gruber F, Serena Leal J, Veltkamp R, Köhrmann M, Berrouschot J. Granulocyte colony-stimulating factor in patients with acute ischemic stroke: results of the AX200 for Ischemic Stroke trial. Stroke 2013; 44:2681-7. [PMID: 23963331 DOI: 10.1161/strokeaha.113.001531] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Granulocyte colony-stimulating factor (G-CSF; AX200; Filgrastim) is a stroke drug candidate with excellent preclinical evidence for efficacy. A previous phase IIa dose-escalation study suggested potential efficacy in humans. The present large phase IIb trial was powered to detect clinical efficacy in acute ischemic stroke patients. METHODS G-CSF (135 µg/kg body weight intravenous over 72 hours) was tested against placebo in 328 patients in a multinational, multicenter, randomized, and placebo-controlled trial (NCT00927836; www.clinicaltrial.gov). Main inclusion criteria were ≤9-hour time window after stroke onset, infarct localization in the middle cerebral artery territory, baseline National Institutes of Health Stroke Scale score range of 6 to 22, and baseline diffusion-weighted imaging lesion size ≥15 mL. Primary and secondary end points were the modified Rankin scale score and the National Institutes of Health Stroke Scale score at day 90, respectively. Data were analyzed using a prespecified model that adjusted for age, National Institutes of Health Stroke Scale score at baseline, and initial infarct volume (diffusion-weighted imaging). RESULTS G-CSF treatment failed to meet the primary and secondary end points of the trial. For additional end points such as mortality, Barthel index, or infarct size at day 30, G-CSF did not show efficacy either. There was, however, a trend for reduced infarct growth in the G-CSF group. G-CSF showed the expected peripheral pharmacokinetic and pharmacodynamic profiles, with a strong increase in leukocytes and monocytes. In parallel, the cytokine profile showed a significant decrease of interleukin-1. CONCLUSIONS G-CSF, a novel and promising drug candidate with a comprehensive preclinical and clinical package, did not provide any significant benefit with respect to either clinical outcome or imaging biomarkers. CLINICAL TRIAL REGISTRATION URL http://www.clinicaltrials.gov. Unique identifier: NCT00927836.
Collapse
Affiliation(s)
- E Bernd Ringelstein
- From the Department of Neurology, University of Münster, Münster, Germany (E.B.R.); Department of Neurology, University of Leuven, Leuven, Belgium (V.T.); Vesalius Research Center, VIB, Leuven, Belgium (V.T.); Department of Clinical Sciences, Section of Neurology, Lund University, Lund, Sweden (B.N.); Department of Neurology, University of Barcelona, Barcelona, Spain (A.C.); Department of Neurology, Nervenklinik Wagner-Jauregg, Linz, Austria (F.A.); Department of Neurology, Kreiskrankenhaus Siegen, Siegen, Germany (M.G.); Department of Neurology, University of California, Los Angeles (J.S.); Clinical Research Department, SYGNIS Bioscience GmbH, Heidelberg, Germany (R.L., A.S., F.R., G.V., G.C.); Center for Stroke Research, Charité, Berlin, Germany (J.B.F.); Department of Neurology, Evangelisches Krankenhaus Bielefeld, Bielefeld, Germany (W.-R.S.); Department of Neurology, UMass Memorial Medical Center, Worcester, MA (M.F.); Department of Neurology, Medical Faculty Hospital Nitra, Nitra, Slovakia (M.B.); Department of Neurology, University Ostrava, Ostrava, Czech Republic (D.S.); Department of Neurology, Allgemeines Krankenhaus Linz, Linz, Austria (F.G.); Department of Neurology, Hospital Universitario Dr Josep Trueta de Girona, Girona, Spain (J.S.L.); Department of Neurology, University of Heidelberg, Heidelberg, Germany (R.V.); Department of Neurology, University of Erlangen, Erlangen, Germany (S.S., R.K., M.K.); and Department of Neurologie, Klinikum Altenburger Land GmbH, Altenburg, Germany (J.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Schmidt A, Minnerup J, Kleinschnitz C. Emerging neuroprotective drugs for the treatment of acute ischaemic stroke. Expert Opin Emerg Drugs 2013; 18:109-20. [DOI: 10.1517/14728214.2013.790363] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Wei RL, Teng HJ, Yin B, Xu Y, Du Y, He FP, Chu KT, Luo BY, Zheng GQ. A systematic review and meta-analysis of buyang huanwu decoction in animal model of focal cerebral ischemia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:138484. [PMID: 23861695 PMCID: PMC3687599 DOI: 10.1155/2013/138484] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/20/2013] [Indexed: 12/04/2022]
Abstract
Buyang Huanwu Decoction (BHD) is a well-known Chinese herbal prescription for ischemic stroke. The objective of this systematic review and meta-analysis is to provide the current evidence for neuroprotective effects of BHD and its possible mechanisms in animal models of focal ischemia. A systematic literature search, through October 2012, was performed using six databases. The outcome measures assessed were infarct size and/or neurological score. Fifty-six studies with 1270 animals that met the inclusion criteria were identified. The median score for methodological quality was 3 with a range of 2 to 6. Compared with vehicle or no treatment controls, BHD gave a 37% improvement in outcome for all doses ranging from 1.0 g/kg to 60 g/kg at each time point that BHD was administered (P < 0.01). Efficacy was higher in mouse models that utilized suture occlusion and temporary ischemia. The neuroprotective effects of BHD are involved in multiple mechanisms and act upon multiple cell types. In conclusion, BHD possesses substantial neuroprotective effects in experimental stroke probably as a result of the multitarget therapy strategy typically utilized in traditional Chinese medicine. Future research should examine the presence of possible experimental bias and an in-depth study of herbal compound preparations.
Collapse
Affiliation(s)
- Rui-li Wei
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Hai-juan Teng
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Bo Yin
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Yang Xu
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Yue Du
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Fang-pin He
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Ke-tan Chu
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
| | - Ben-yan Luo
- Brain Medical Center, First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou Zhejiang 31000, China
- *Ben-yan Luo: and
| | - Guo-qing Zheng
- The Center of Neurology and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical College, Wenzhou 325027, China
- *Guo-qing Zheng:
| |
Collapse
|
50
|
Liu J, Zhang C, Tao W, Liu M. Systematic review and meta-analysis of the efficacy of sphingosine-1-phosphate (S1P) receptor agonist FTY720 (fingolimod) in animal models of stroke. Int J Neurosci 2012; 123:163-9. [PMID: 23167788 DOI: 10.3109/00207454.2012.749255] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE FTY720 (fingolimod) is a known sphingosine-1-phosphate (S1P) receptor agonist, which has been used in clinical trials for treating multiple sclerosis, renal transplantation, and decreasing reperfusion injury in heart, liver, and kidney. Most of these clinical trials have showed a positive effect. Especially, the trials of MS showed a reduction of relapse rate in FTY720-treated patients. Now, some animal experiments indicated that FTY720 could be a new compound available treatment for stroke patients by exerting neuroprotection via S1P1 mediated antiapoptotic mechanisms. Whether it could be effective in animals is unclear, so we conducted a systematic review to make it clear. METHODS We conducted a systematic review and meta-analysis of the efficacy of FTY720 in animal models of focal cerebral ischemia by electronic and manual searches of the literature. Data on study quality, FTY720 dose, time of administration, and outcome measured as infarct volume or functional deficit were extracted. Data from all studies were analyzed by means of a standardized mean difference meta-analysis. RESULTS Of the 19 identified studies, 9 were included. Among all the included studies, 178 animals were calculated for infarct size and 194 animals were assessed of neurological deficits. The methodological quality of the studies ranged from 2 to 10 according to a published 11-item quality scale. Of the nine studies selected, only one reported a negative result of FTY720. The result indicated that FTY720 reduced the infarct volume (SMD = -1.31, 95% CI -1.99 to -0.63) and improve the functional outcome (SMD = -1.61, 95% CI -2.17 to -1.05). CONCLUSIONS The data we included supporting FTY720 was a candidate drug for stroke, but it should be considered with caution. More good quality experimental studies should be performed to evaluate the safety of FTY720 in the future. Whether FTY720 is effective in aged animals that mimicked human with comorbidities like diabetes and hypertension should also be deliberated.
Collapse
Affiliation(s)
- Junfeng Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | |
Collapse
|