1
|
Wang Y, Yu Z, Cheng M, Hu E, Yan Q, Zheng F, Guo X, Zhang W, Li H, Li Z, Zhu W, Wu Y, Tang T, Li T. Buyang huanwu decoction promotes remyelination via miR-760-3p/GPR17 axis after intracerebral hemorrhage. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118126. [PMID: 38556140 DOI: 10.1016/j.jep.2024.118126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/02/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The repairment of myelin sheaths is crucial for mitigating neurological impairments of intracerebral hemorrhage (ICH). However, the current research on remyelination processes in ICH remains limited. A representative traditional Chinese medicine, Buyang Huanwu decoction (BYHWD), shows a promising therapeutic strategy for ICH treatment. AIM OF THE STUDY To investigate the pro-remyelination effects of BYHWD on ICH and explore the underlying mechanisms. MATERIALS AND METHODS The collagenase-induced mice ICH model was created for investigation. BYHWD's protective effects were assessed by behavioral tests and histological staining. Transmission electron microscopy was used for displaying the structure of myelin sheaths. The remyelination and oligodendrocyte differentiation were evaluated by the expressions of myelin proteolipid protein (PLP), myelin basic protein (MBP), MBP/TAU, Olig2/CC1, and PDGFRα/proliferating cell nuclear antigen (PCNA) through RT-qPCR and immunofluorescence. Transcriptomics integrated with disease database analysis and experiments in vivo and in vitro revealed the microRNA-related underlying mechanisms. RESULTS Here, we reported that BYHWD promoted the neurological function of ICH mice and improved remyelination by increasing PLP, MBP, and TAU, as well as restoring myelin structure. Besides, we showed that BYHWD promoted remyelination by boosting the differentiation of PDGFRα+ oligodendrocyte precursor cells into olig2+/CC1+ oligodendrocytes. Additionally, we demonstrated that the remyelination effects of BYHWD worked by inhibiting G protein-coupled receptor 17 (GPR17). miRNA sequencing integrated with miRNA database prediction screened potential miRNAs targeting GPR17. By applying immunofluorescence, RNA in situ hybridization and dual luciferase reporter gene assay, we confirmed that BYHWD suppressed GPR17 and improved remyelination by increasing miR-760-3p. CONCLUSIONS BYHWD improves remyelination and neurological function in ICH mice by targeting miR-760-3p to inhibit GPR17. This study may shed light on the orchestration of remyelination mechanisms after ICH, thus providing novel insights for developing innovative prescriptions with brain-protective properties.
Collapse
Affiliation(s)
- Yang Wang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China
| | - Zhe Yu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Menghan Cheng
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - En Hu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China
| | - Qiuju Yan
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Fei Zheng
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Xiaohang Guo
- School of Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, PR China
| | - Haigang Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan, PR China
| | - Zhilin Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Wenxin Zhu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yao Wu
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Tao Tang
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China
| | - Teng Li
- Institute of Integrative Medicine, Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; NATCM Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Xiangya Hospital, Central South University, Jiangxi, Nanchang, PR China.
| |
Collapse
|
2
|
Bone M, Malik M, Crilly S. Identifying applications of virtual reality to benefit the stroke translational pipeline. Brain Neurosci Adv 2023; 7:23982128231182506. [PMID: 37360628 PMCID: PMC10288399 DOI: 10.1177/23982128231182506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
As a leading cause of mortality and morbidity, stroke and its management have been studied extensively. Despite numerous pre-clinical studies identifying therapeutic targets, development of effective, specific pharmacotherapeutics remain limited. One significant limitation is a break in the translational pipeline - promising pre-clinical results have not always proven replicable in the clinic. Recent developments in virtual reality technology might help generate a better understanding of injury and recovery across the whole research pipeline in search of optimal stroke management. Here, we review the technologies that can be applied both clinically and pre-clinically to stroke research. We discuss how virtual reality technology is used to quantify clinical outcomes in other neurological conditions that have potential to be applied in stroke research. We also review current uses in stroke rehabilitation and suggest how immersive programmes would better facilitate the quantification of stroke injury severity and patient recovery comparable to pre-clinical study design. By generating continuous, standardised and quantifiable data from injury onset to rehabilitation, we propose that by paralleling pre-clinical outcomes, we can apply a better reverse-translational strategy and apply this understanding to animal studies. We hypothesise this combination of translational research strategies may improve the reliability of pre-clinical research outcomes and culminate in real-life translation of stroke management regimens and medications.
Collapse
Affiliation(s)
- Matan Bone
- School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre and The University of Manchester, Manchester, UK
| | - Maham Malik
- School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre and The University of Manchester, Manchester, UK
| | - Siobhan Crilly
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre and The University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and The University of Manchester, Manchester, UK
| |
Collapse
|
3
|
Peterson C, Hawk C, Puglisi CH, Waldau B. Intracranial Pressure Monitoring In Nontraumatic Intraventricular Hemorrhage Rodent Model. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2022:10.3791/63309. [PMID: 35225289 PMCID: PMC9481275 DOI: 10.3791/63309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Survivors of intraventricular hemorrhage are often left with significant long-term memory impairment; thus, research utilizing intraventricular hemorrhage animal models is essential. In this study, we sought out ways to measure intracranial pressure, mean arterial pressure, and cerebral perfusion pressure during nontraumatic intraventricular hemorrhage in rodents. The experimental design included three Sprague Dawley groups: sham, standard 200 µl intraventricular hemorrhage, and vehicle control groups. By introducing an intraparenchymal fiberoptic pressure sensor, precise intracranial pressure measurements were obtained in all groups. Cerebral perfusion pressures were calculated with the knowledge of intracranial pressure and mean arterial pressure values. As expected, the intraventricular hemorrhage and vehicle control groups both experienced a rise in the intracranial pressure and subsequent decline in cerebral perfusion pressure during intraventricular injection of autologous blood and artificial cerebrospinal fluid, respectively. The addition of an intraparenchymal fiberoptic pressure sensor is beneficial in monitoring precise intracranial pressure changes.
Collapse
Affiliation(s)
| | - Cameron Hawk
- Department of Neurological Surgery, UC Davis Medical Center
| | | | - Ben Waldau
- Department of Neurological Surgery, UC Davis Medical Center
| |
Collapse
|
4
|
Aronowski J, Sansing LH, Xi G, Zhang JH. Mechanisms of Damage After Cerebral Hemorrhage. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
5
|
Peterson C, Umoye AO, Puglisi CH, Waldau B. Mechanisms of memory impairment in animal models of nontraumatic intracranial hemorrhage: A systematic review of the literature. BRAIN HEMORRHAGES 2021; 3:77-93. [DOI: 10.1016/j.hest.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
6
|
Wilkinson CM, Kung TF, Jickling GC, Colbourne F. A translational perspective on intracranial pressure responses following intracerebral hemorrhage in animal models. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2020.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
7
|
Liddle LJ, Ralhan S, Ward DL, Colbourne F. Translational Intracerebral Hemorrhage Research: Has Current Neuroprotection Research ARRIVEd at a Standard for Experimental Design and Reporting? Transl Stroke Res 2020; 11:1203-1213. [PMID: 32504197 PMCID: PMC7575495 DOI: 10.1007/s12975-020-00824-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/17/2023]
Abstract
One major aim of preclinical intracerebral hemorrhage (ICH) research is to develop and test potential neuroprotectants. Published guidelines for experimental design and reporting stress the importance of clearly and completely reporting results and methodological details to ensure reproducibility and maximize information availability. The current review has two objectives: first, to characterize current ICH neuroprotection research and, second, to analyze aspects of translational design in preclinical ICH studies. Translational design is the adoption and reporting of experimental design characteristics that are thought to be clinically relevant and critical to reproducibility in animal studies (e.g., conducting and reporting experiments according to the STAIR and ARRIVE guidelines, respectively). Given that ICH has no current neuroprotective treatments and an ongoing reproducibility crisis in preclinical research, translational design should be considered by investigators. We conducted a systematic review of ICH research from 2015 to 2019 using the PubMed database. Our search returned 281 published manuscripts studying putative neuroprotectants in animal models. Contemporary ICH research predominantly uses young, healthy male rodents. The collagenase model is the most commonly used. Reporting of group sizes, blinding, and randomization are almost unanimous, but group size calculations, mortality and exclusion criteria, and animal model characteristics are infrequently reported. Overall, current ICH neuroprotection research somewhat aligns with experimental design and reporting guidelines. However, there are areas for improvement. Because failure to consider translational design is associated with inflation of effect sizes (and possibly hindered reproducibility), we suggest that researchers, editors, and publishers collaboratively consider enhanced adherence to published guidelines.
Collapse
Affiliation(s)
- Lane J Liddle
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Shivani Ralhan
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel L Ward
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada.
| |
Collapse
|
8
|
Martins CA, Neves LT, de Oliveira MMBP, Bagatini PB, Barboza R, Mestriner RG, Xavier LL, Rasia-Filho AA. Neuroprotective effect of ACTH on collagenase-induced peri-intraventricular hemorrhage in newborn male rats. Sci Rep 2020; 10:17734. [PMID: 33082383 PMCID: PMC7576182 DOI: 10.1038/s41598-020-74712-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 10/06/2020] [Indexed: 12/23/2022] Open
Abstract
Peri-intraventricular hemorrhage (PIVH) is a common and serious prematurity-related complication in neonates. Adrenocorticotropic hormone (ACTH) has neuroprotective actions and is a candidate to ameliorate brain damage following PIVH. Here, we tested the efficacy of ACTH1-24 on a collagenase-induced lesion of the germinal matrix (GM) in newborn male rats. Animals received microinjection of the vehicle (PBS, 2 µl) or collagenase type VII (0.3 IU) into the GM/periventricular tissue on postnatal day (PN) 2. Twelve hours later pups received microinjection of either the agonist ACTH1-24 (0.048 mg/kg), or the antagonist SHU9119 (antagonist of MCR3/MCR4 receptors, 0.01 mg/kg), or their combination. Morphological outcomes included striatal injury extension, neuronal and glial cells counting, and immunohistochemical expression of brain lesion biomarkers ipsilateral and contralateral to the hemorrhagic site. Data were evaluated on PN 8. Collagenase induced PIVH and severe ipsilateral striatal lesion. ACTH1-24 dampened the deleterious effects of collagenase-induced hemorrhage in significantly reducing the extension of the damaged area, the striatal neuronal and glial losses, and the immunoreactive expression of the GFAP, S100β, and NG2-glia biomarkers in the affected periventricular area. SHU9119 blocked the glial density rescuing effect of ACTH1-24. ACTH1-24 could be further evaluated to determine its suitability for preclinical models of PVH in premature infants.
Collapse
Affiliation(s)
- Camila A Martins
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, 90170-050, Brazil
- Departamento de Ciências Básicas da Saúde/Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, R. Sarmento Leite 245, Porto Alegre, RS, 90170-050, Brazil
| | - Laura Tartari Neves
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Marina M B P de Oliveira
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Pamela Brambilla Bagatini
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Rafaela Barboza
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Régis Gemerasca Mestriner
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Léder Leal Xavier
- Laboratório de Biologia Celular e Tecidual, Escola de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, 90619-900, Brazil
| | - Alberto A Rasia-Filho
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, 90170-050, Brazil.
- Departamento de Ciências Básicas da Saúde/Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, R. Sarmento Leite 245, Porto Alegre, RS, 90170-050, Brazil.
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90170-050, Brazil.
| |
Collapse
|
9
|
|
10
|
Mello TG, Rosado-de-Castro PH, Campos RMP, Vasques JF, Rangel-Junior WS, Mattos RSDARD, Puig-Pijuan T, Foerster BU, Gutfilen B, Souza SAL, Boltze J, Paiva FF, Mendez-Otero R, Pimentel-Coelho PM. Intravenous Human Umbilical Cord-Derived Mesenchymal Stromal Cell Administration in Models of Moderate and Severe Intracerebral Hemorrhage. Stem Cells Dev 2020; 29:586-598. [PMID: 32160799 DOI: 10.1089/scd.2019.0176] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is as a life-threatening condition that can occur in young adults, often causing long-term disability. Recent preclinical data suggest mesenchymal stromal cell (MSC)-based therapies as promising options to minimize brain damage after ICH. However, therapeutic evidence and mechanistic insights are still limited, particularly when compared with other disorders such as ischemic stroke. Herein, we employed a model of collagenase-induced ICH in young adult rats to investigate the potential therapeutic effects of an intravenous injection of human umbilical cord Wharton's jelly-derived MSCs (hUC-MSCs). Two doses of collagenase were used to cause moderate or severe hemorrhages. Magnetic resonance imaging showed that animals treated with hUC-MSCs after moderate ICH had smaller residual hematoma volumes than vehicle-treated rats, whereas the cell therapy failed to decrease the hematoma volume in animals with a severe ICH. Functional assessments (rotarod and elevated body swing tests) were performed for up to 21 days after ICH. Enduring neurological impairments were seen only in animals subjected to severe ICH, but the cell therapy did not induce statistically significant improvements in the functional recovery. The biodistribution of Technetium-99m-labeled hUC-MSCs was also evaluated, showing that most cells were found in organs such as the spleen and lungs 24 h after transplantation. Nevertheless, it was possible to detect a weak signal in the brain, which was higher in the ipsilateral hemisphere of rats subjected to a severe ICH. These data indicate that hUC-MSCs have moderately beneficial effects in cases of less severe brain hemorrhages in rats by decreasing the residual hematoma volume, and that optimization of the therapy is still necessary.
Collapse
Affiliation(s)
- Tanira Giara Mello
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Engenharia Nuclear, Comissão Nacional de Energia Nuclear, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Paulo Henrique Rosado-de-Castro
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil.,Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Juliana Ferreira Vasques
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | | | | | - Teresa Puig-Pijuan
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Bernd Uwe Foerster
- Instituto de Física de São Carlos, Universidade de São Paulo, São Carlos, Brazil
| | - Bianca Gutfilen
- Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio Augusto Lopes Souza
- Departamento de Radiologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | | | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| | - Pedro Moreno Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Grüter BE, Croci D, Schöpf S, Nevzati E, d’Allonzo D, Lattmann J, Roth T, Bircher B, Muroi C, Dutilh G, Widmer HR, Plesnila N, Fandino J, Marbacher S. Systematic Review and Meta-analysis of Methodological Quality in In Vivo Animal Studies of Subarachnoid Hemorrhage. Transl Stroke Res 2020; 11:1175-1184. [DOI: 10.1007/s12975-020-00801-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023]
|
12
|
Wilkinson CM, Kalisvaart ACJ, Kung TFC, Maisey DR, Klahr AC, Dickson CT, Colbourne F. The collagenase model of intracerebral hemorrhage in awake, freely moving animals: The effects of isoflurane. Brain Res 2019; 1728:146593. [PMID: 31816320 DOI: 10.1016/j.brainres.2019.146593] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/13/2019] [Accepted: 12/04/2019] [Indexed: 01/17/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating stroke often modelled in rats. Isoflurane anesthetic, commonly used in preclinical research, affects general physiology (e.g., blood pressure) and electrophysiology (e.g., burst suppression) in many ways. These physiological changes may detract from the clinical relevance of the model. Here, we revised the standard collagenase model to produce an ICH in rats without anesthetic. Guide cannulas were implanted stereotaxically under anesthetic. After 3 days of recovery, collagenase was infused through an internal cannula into the striatum of animals randomly assigned to the non-anesthetized or isoflurane group. We assessed whether isoflurane affected hematoma volume, core temperature, movement activity, pain, blood pressure, and seizure activity. With a small ICH, there was a hematoma volume increased from 8.6 (±3.3, 95% confidence interval) µL in anesthetized rats to 13.2 (±3.1) µL in non-anesthetized rats (P = 0.008), but with a larger ICH, hematoma volumes were similar. Isoflurane decreased temperature by 1.3 °C (±0.16 °C, P < 0.001) for 2 h and caused a 35.1 (±1.7) mmHg group difference in blood pressure (P < 0.007) for 12 m. Blood glucose increased twofold after isoflurane procedures (P < 0.001). Pain, as assessed with the rat grimace scale, did not differ between groups. Seizure incidence rate (62.5%) in non-anesthetized ICH rats was similar to historic amounts (61.3%). In conclusion, isoflurane appears to have some significant and injury size-dependent effects on the collagenase model. Thus, when anesthetic effects are a known concern, the use of the standardized cannula infusion approach is scientifically and ethically acceptable.
Collapse
Affiliation(s)
| | | | - Tiffany F C Kung
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - D Ryan Maisey
- Social Sciences - Augustana Faculty, University of Alberta, Camrose, Canada
| | - Ana C Klahr
- Social Sciences - Augustana Faculty, University of Alberta, Camrose, Canada
| | - Clayton T Dickson
- Department of Psychology, University of Alberta, Edmonton, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
13
|
Glibenclamide, a Sur1-Trpm4 antagonist, does not improve outcome after collagenase-induced intracerebral hemorrhage. PLoS One 2019; 14:e0215952. [PMID: 31042750 PMCID: PMC6494051 DOI: 10.1371/journal.pone.0215952] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/11/2019] [Indexed: 12/16/2022] Open
Abstract
The sulfonylurea 1 transient receptor potential melastatin 4 (Sur1-Trpm4) receptor is selectively expressed after intracerebral hemorrhage (ICH). This upregulation contributes to increases in intracellular sodium. Water follows sodium through aquaporin channels, leading to cytotoxic edema. Even after edema is thought to have resolved, ionic dyshomeostasis persists, as does blood-brain barrier (BBB) damage. Glibenclamide, a hypoglycemic agent that inhibits Sur1-Trpm4, has been shown to reduce BBB damage and edema following infusion of autologous blood into the brain (ICH) as well as after other brain injuries. In order to further assess efficacy, we used the collagenase ICH model in rats to test whether glibenclamide reduces edema, attenuates ion dyshomeostasis, improves BBB damage, and reduces lesion volume. We tested a widely-used glibenclamide dose shown effective in other studies (10 μg/kg loading dose followed by 200 ng/hr for up to 7 days). Early initiation of glibenclamide did not significantly impact edema (72 hours), BBB permeability (72 hours), or lesion volume after ICH (28 days). Recovery from neurological impairments was also not improved by glibenclamide. These results suggest that glibenclamide will not improve outcome in ICH. However, the treatment appeared to be safe as there was no effect on bleeding or other physiological variables.
Collapse
|
14
|
Liu Y, Lu G, Su XW, Ding T, Wang WL, Li YM, Poon WS, Ao LJ. Characterization of Axon Damage, Neurological Deficits, and Histopathology in Two Experimental Models of Intracerebral Hemorrhage. Front Neurosci 2018; 12:928. [PMID: 30618557 PMCID: PMC6297275 DOI: 10.3389/fnins.2018.00928] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/26/2018] [Indexed: 12/22/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) is one of the most lethal forms of stroke. From the limited previous studies and our preliminary data, white matter is considered a key predictor of the outcome and potential target of recovery. The traditional ICH model induced by injection of autologous blood or bacterial collagenase into striatum (ST) demonstrated a spontaneous functional recovery within one or 2 months. We hypothesis that an internal capsule (IC) lesion might lead to long-term axonal damage and long lasting functional deficits. Thus in this study, a modified internal capsule ICH model was conducted in rats, and the axonal damage, neurological deficits, histopathology as well as electrophysiology were characterized. The finding demonstrated that compared to ST group, the modified IC lesioned model exhibited a relatively smaller lesion volume with consistent axonal loss/degeneration and long-lasting neurological dysfunction at 2 months after ICH. Functionally, the impairment of the mNSS, ratio of contralateral forelimb usage, four limb stand index, contralateral duty cycle and ipsilateral SSEPs amplitude remained significant at 56 days. Structurally, the significant loss of PKCγ in ipsilateral cortical spinal tracts of IC group and the consistent axonal degeneration with several axonal retraction bulbs and enlarged tubular space was observed at 56 days after ICH. This study suggested that a modified IC lesioned model was proved to have long lasting neurological deficits. A comprehensive understanding of the dynamic progression after experimental ICH should aid further successful clinic translation in animal ICH studies, and provide new insights into the role of whiter matter injury in the mechanism and therapeutic targets of ICH.
Collapse
Affiliation(s)
- Yao Liu
- School of Rehabilitation, Kunming Medical University, Kunming, China.,Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Gang Lu
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xian Wei Su
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tao Ding
- Rehabilitation Department, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen Li Wang
- Rehabilitation Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Mei Li
- Rehabilitation Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Li Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, China
| |
Collapse
|
15
|
Li W, Li R, Zhao S, Jiang C, Liu Z, Tang X. Lithium Posttreatment Alleviates Blood–Brain Barrier Injury After Intracerebral Hemorrhage in Rats. Neuroscience 2018; 383:129-137. [DOI: 10.1016/j.neuroscience.2018.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/02/2018] [Accepted: 05/01/2018] [Indexed: 10/16/2022]
|
16
|
Wilkinson DA, Keep RF, Hua Y, Xi G. Hematoma clearance as a therapeutic target in intracerebral hemorrhage: From macro to micro. J Cereb Blood Flow Metab 2018; 38:741-745. [PMID: 29350086 PMCID: PMC5888862 DOI: 10.1177/0271678x17753590] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite the absence of an intervention shown to improve outcomes in intracerebral hemorrhage, preclinical work has led to a greater understanding of the pathologic pathways of brain injury. Methods targeting hematoma clearance through both macroscopic (surgical) and microscopic (endogenous phagocytosis) means are currently under investigation, with multiple clinical trials ongoing. Macroscopic methods for removal involve both catheter- and endoscope-based therapies to remove the hematoma through minimally invasive surgery. Microscopic methods targeting hematoma clearance involve augmenting endogenous clearance pathways for red blood cells and altering the balance between phagocytosis and red blood cell lysis with the release of potentially harmful constituents (e.g. hemoglobin and iron) into the extracellular space.
Collapse
Affiliation(s)
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
17
|
Katsuki H, Hijioka M. Intracerebral Hemorrhage as an Axonal Tract Injury Disorder with Inflammatory Reactions. Biol Pharm Bull 2018; 40:564-568. [PMID: 28458342 DOI: 10.1248/bpb.b16-01013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a neurological disorder frequently accompanied by severe dysfunction. Critical pathogenic events leading to poor prognosis should be identified for the development of novel effective therapies for ICH. Here we focus on the injury of the axonal tract, particularly of the internal capsule, with reference to its contribution to ICH pathology and potential therapeutic interventions in addition to its cellular mechanisms. Studies on human ICH patients and rodent models of ICH suggest that invasion of hematoma into the internal capsule greatly worsens the severity of post-ICH symptoms. A blood-derived protease thrombin may play an important role in the acute phase of axonal tract injury in the internal capsule that includes compromised axonal transport and fragmentation of axonal structures. Several agents such as clioquinol, melatonin and Am80 (a retinoic acid receptor agonist) have been shown to produce therapeutic effects on rodent models of ICH associated with injury of the internal capsule. In the course of examinations on the effect of Am80, we obtained evidence for the involvement of CXCL2, a neutrophil chemotactic factor, in the pathogenesis of ICH. Accordingly, we also refer to the potential roles of infiltrating neutrophils and inflammatory responses in axonal tract injury and resultant neurological dysfunction in ICH.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Masanori Hijioka
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
18
|
Li Q, Wan J, Lan X, Han X, Wang Z, Wang J. Neuroprotection of brain-permeable iron chelator VK-28 against intracerebral hemorrhage in mice. J Cereb Blood Flow Metab 2017; 37:3110-3123. [PMID: 28534662 PMCID: PMC5584702 DOI: 10.1177/0271678x17709186] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/14/2017] [Accepted: 04/18/2017] [Indexed: 01/08/2023]
Abstract
Iron overload plays a key role in the secondary brain damage that develops after intracerebral hemorrhage (ICH). The significant increase in iron deposition is associated with the generation of reactive oxygen species (ROS), which leads to oxidative brain damage. In this study, we examined the protective effects of VK-28, a brain-permeable iron chelator, against hemoglobin toxicity in an ex vivo organotypic hippocampal slice culture (OHSC) model and in middle-aged mice subjected to an in vivo, collagenase-induced ICH model. We found that the effects of VK-28 were similar to those of deferoxamine (DFX), a well-studied iron chelator. Both decreased cell death and ROS production in OHSCs and in vivo, decreased iron-deposition and microglial activation around hematoma in vivo, and improved neurologic function. Moreover, compared with DFX, VK-28 polarized microglia to an M2-like phenotype, reduced brain water content, deceased white matter injury, improved neurobehavioral performance, and reduced overall death rate after ICH. The protection of VK-28 was confirmed in a blood-injection ICH model and in aged-male and young female mice. Our findings indicate that VK-28 is protective against iron toxicity after ICH and that, at the dosage tested, it has better efficacy and less toxicity than DFX does.
Collapse
Affiliation(s)
| | | | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhongyu Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Williamson MR, Colbourne F. Evidence for Decreased Brain Parenchymal Volume After Large Intracerebral Hemorrhages: a Potential Mechanism Limiting Intracranial Pressure Rises. Transl Stroke Res 2017; 8:386-396. [PMID: 28281221 PMCID: PMC5493716 DOI: 10.1007/s12975-017-0530-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/26/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022]
Abstract
Potentially fatal intracranial pressure (ICP) rises commonly occur after large intracerebral hemorrhages (ICH). We monitored ICP after infusing 100-160 μL of autologous blood (vs. 0 μL control) into the striatum of rats in order to test the validity of this common model with regard to ICP elevations. Other endpoints included body temperature, behavioral impairment, lesion volume, and edema. Also, we evaluated hippocampal CA1 sector and somatosensory cortical neuron morphology to assess whether global ischemic injury occurred. Despite massive blood infusions, ICP only modestly increased (160 μL 10.8 ± 2.1 mmHg for <36 h vs. control 3.4 ± 0.5 mmHg), with little peri-hematoma edema at 3 days. Body temperature was not affected. Behavioral deficits and tissue loss were infusion volume-dependent. There was no histological evidence of hippocampal or cortical injury, indicating that cell death was confined to the hematoma and closely surrounding tissue. Surprisingly, the most severe hemorrhages significantly increased cell density (~15-20%) and reduced cell body size (~30%) in regions outside the injury site. Additionally, decreased cell size and increased density were observed after collagenase-induced ICH. Parenchymal volume is seemingly reduced after large ICH. Thus, in addition to well-known compliance mechanisms (e.g., displacement of cerebrospinal fluid and cerebral blood), reduced brain parenchymal volume appears to limit ICP rises in rodents with very large mass lesions.
Collapse
Affiliation(s)
- Michael R Williamson
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, Alberta, T6G 2E9, Canada.
| |
Collapse
|
20
|
Wowk S, Fagan KJ, Ma Y, Nichol H, Colbourne F. Examining potential side effects of therapeutic hypothermia in experimental intracerebral hemorrhage. J Cereb Blood Flow Metab 2017; 37:2975-2986. [PMID: 27899766 PMCID: PMC5536807 DOI: 10.1177/0271678x16681312] [Citation(s) in RCA: 7] [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: 07/11/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 11/16/2022]
Abstract
Studies treating intracerebral hemorrhage (ICH) with therapeutic hypothermia (TH) have shown inconsistent benefits. We hypothesized that TH's anti-inflammatory effects may be responsible as inflammatory cells are essential for removing degrading erythrocytes. Here, we subjected rats to a collagenase-induced striatal ICH followed by whole-body TH (∼33℃ for 11-72 h) or normothermia. We used X-ray fluorescence imaging to spatially quantify total and peri-hematoma iron three days post-injury. At three and seven days, we measured non-heme iron levels. Finally, hematoma volume was quantified on one, three, and seven days. In the injured hemisphere, total iron levels were elevated ( p < 0.001) with iron increasing in the peri-hematoma region ( p = 0.007). Non-heme iron increased from three to seven days (p < 0.001). TH had no effect on any measure of iron ( p ≥ 0.479). At one and three days, TH did not affect hematoma volume ( p ≥ 0.264); however, at seven days there was a four-fold increase in hematoma volume in 40% of treated animals ( p = 0.032). Thus, even when TH does not interfere with initial increases in total and non-heme iron or its containment, TH can cause re-bleeding post-treatment. This serious complication could partly account for the intermittent protection previously observed. This also raises serious concerns for clinical usage of TH for ICH.
Collapse
Affiliation(s)
- Shannon Wowk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kelly J Fagan
- Department of Biology, MacEwan University, Edmonton, Canada
| | - Yonglie Ma
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Helen Nichol
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Lan X, Han X, Li Q, Yang QW, Wang J. Modulators of microglial activation and polarization after intracerebral haemorrhage. Nat Rev Neurol 2017; 13:420-433. [PMID: 28524175 PMCID: PMC5575938 DOI: 10.1038/nrneurol.2017.69] [Citation(s) in RCA: 553] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral haemorrhage (ICH) is the most lethal subtype of stroke but currently lacks effective treatment. Microglia are among the first non-neuronal cells on the scene during the innate immune response to ICH. Microglia respond to acute brain injury by becoming activated and developing classic M1-like (proinflammatory) or alternative M2-like (anti-inflammatory) phenotypes. This polarization implies as yet unrecognized actions of microglia in ICH pathology and recovery, perhaps involving microglial production of proinflammatory or anti-inflammatory cytokines and chemokines. Furthermore, alternatively activated M2-like microglia might promote phagocytosis of red blood cells and tissue debris, a major contribution to haematoma clearance. Interactions between microglia and other cells modulate microglial activation and function, and are also important in ICH pathology. This Review summarizes key studies on modulators of microglial activation and polarization after ICH, including M1-like and M2-like microglial phenotype markers, transcription factors and key signalling pathways. Microglial phagocytosis, haematoma resolution, and the potential crosstalk between microglia and T lymphocytes, neurons, astrocytes, and oligodendrocytes in the ICH brain are described. Finally, the clinical and translational implications of microglial polarization in ICH are presented, including the evidence that therapeutic approaches aimed at modulating microglial function might mitigate ICH injury and improve brain repair.
Collapse
Affiliation(s)
- Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Qian Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| |
Collapse
|
22
|
Karuppagounder SS, Alim I, Khim SJ, Bourassa MW, Sleiman SF, John R, Thinnes CC, Yeh TL, Demetriades M, Neitemeier S, Cruz D, Gazaryan I, Killilea DW, Morgenstern L, Xi G, Keep RF, Schallert T, Tappero RV, Zhong J, Cho S, Maxfield FR, Holman TR, Culmsee C, Fong GH, Su Y, Ming GL, Song H, Cave JW, Schofield CJ, Colbourne F, Coppola G, Ratan RR. Therapeutic targeting of oxygen-sensing prolyl hydroxylases abrogates ATF4-dependent neuronal death and improves outcomes after brain hemorrhage in several rodent models. Sci Transl Med 2016; 8:328ra29. [PMID: 26936506 DOI: 10.1126/scitranslmed.aac6008] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier-permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.
Collapse
Affiliation(s)
- Saravanan S Karuppagounder
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Ishraq Alim
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Soah J Khim
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Megan W Bourassa
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Sama F Sleiman
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Roseleen John
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | | | - Tzu-Lan Yeh
- Department of Chemistry, University of Oxford, OX1 3TA Oxford, UK
| | | | - Sandra Neitemeier
- Institut fuer Pharmakologie and Klinische Pharmazie, Phillips-Universitaet Marburg, D 35032 Marburg, Germany
| | - Dana Cruz
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Irina Gazaryan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | - Lewis Morgenstern
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Timothy Schallert
- Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
| | - Ryan V Tappero
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Jian Zhong
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Sunghee Cho
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Frederick R Maxfield
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Theodore R Holman
- Chemistry and Biochemistry, Department, University of California at Santa Cruz, Santa Cruz, CA 95064, USA
| | - Carsten Culmsee
- Institut fuer Pharmakologie and Klinische Pharmazie, Phillips-Universitaet Marburg, D 35032 Marburg, Germany
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Yijing Su
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - John W Cave
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Giovanni Coppola
- Department of Psychiatry, University of California at Los Angeles, CA 90095, USA
| | - Rajiv R Ratan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
23
|
Zheng J, Liu Z, Li W, Tang J, Zhang D, Tang X. Lithium posttreatment confers neuroprotection through glycogen synthase kinase-3β inhibition in intracerebral hemorrhage rats. J Neurosurg 2016; 127:716-724. [PMID: 27739937 DOI: 10.3171/2016.7.jns152995] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Inflammation and apoptosis are two key factors contributing to secondary brain injury after intracerebral hemorrhage (ICH). The objective of this study was to evaluate the effects of lithium posttreatment on behavior, brain atrophy, inflammation, and perihematomal cell death. Furthermore, the authors aimed to determine the role of the pro-apoptotic glycogen synthase kinase-3β (GSK-3β) after experimental ICH. METHODS Male Sprague-Dawley rats (n = 108) were subjected to intracerebral infusion of semicoagulated autologous blood. Window of opportunity and dose optimization studies of lithium on ICH-induced injury were performed by measuring neurological deficits. Animals with ICH received vehicle administration or lithium posttreatment (60 mg/kg) for up to 21 days. Hemispheric atrophy was evaluated. Perihematomal cell death was quantified through terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL). The number of myeloperoxidase (MPO)-positive neutrophils and OX42-positive microglia in the perihematomal areas were calculated. Western blotting was used for the quantification of GSK-3β, heat shock protein 70 (HSP70), nuclear factor-κB p65 (NF-κB p65), and cy-clooxygenase-2 (COX-2). RESULTS Lithium, at a dose of 60 mg/kg initiated from 2 hours after injury, exhibited the best effects of improving neurological outcomes 3, 5, 7, 14, 21, and 28 days after ICH, reduced the hemispheric atrophy at 42 days after surgery, and reduced the number of TUNEL-positive cells, MPO-positive neutrophils, and OX42-positive microglia in the perihematomal areas. Furthermore, lithium posttreatment modulated GSK-3β, increased HSP70, and decreased NF-κB p65 and COX-2 expression in the ipsilateral hemisphere. CONCLUSIONS Lithium posttreatment at a dose of 60 mg/kg, initiated beginning 2 hours after injury, improves functional and morphological outcomes, and inhibits inflammation and perihematomal cell death in a rat model of semicoagulated autologous blood ICH through inactivation of GSK-3β.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Heilongjiang
| | - Zhen Liu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Heilongjiang
| | - Weishan Li
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Heilongjiang
| | - Jiaxin Tang
- The Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai; and
| | - Dongwei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang, China
| | - Xiaobo Tang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Heilongjiang
| |
Collapse
|
24
|
Mouse model of intracerebellar haemorrhage. Behav Brain Res 2016; 312:374-84. [DOI: 10.1016/j.bbr.2016.06.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/12/2016] [Accepted: 06/16/2016] [Indexed: 12/14/2022]
|
25
|
Review of Preclinical and Clinical Studies of Bone Marrow-Derived Cell Therapies for Intracerebral Hemorrhage. Stem Cells Int 2016; 2016:4617983. [PMID: 27698671 PMCID: PMC5028871 DOI: 10.1155/2016/4617983] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 06/01/2016] [Indexed: 12/22/2022] Open
Abstract
Stroke is the second leading cause of mortality worldwide, causing millions of deaths annually, and is also a major cause of disability-adjusted life years. Hemorrhagic stroke accounts for approximately 10 to 27% of all cases and has a fatality rate of about 50% in the first 30 days, with limited treatment possibilities. In the past two decades, the therapeutic potential of bone marrow-derived cells (particularly mesenchymal stem cells and mononuclear cells) has been intensively investigated in preclinical models of different neurological diseases, including models of intracerebral hemorrhage and subarachnoid hemorrhage. More recently, clinical studies, most of them small, unblinded, and nonrandomized, have suggested that the therapy with bone marrow-derived cells is safe and feasible in patients with ischemic or hemorrhagic stroke. This review discusses the available evidence on the use of bone marrow-derived cells to treat hemorrhagic strokes. Distinctive properties of animal studies are analyzed, including study design, cell dose, administration route, therapeutic time window, and possible mechanisms of action. Furthermore, clinical trials are also reviewed and discussed, with the objective of improving future studies in the field.
Collapse
|
26
|
Klahr AC, Nadeau CA, Colbourne F. Temperature Control in Rodent Neuroprotection Studies: Methods and Challenges. Ther Hypothermia Temp Manag 2016; 7:42-49. [PMID: 27327871 DOI: 10.1089/ther.2016.0018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Extensive animal research facilitated the clinical translation of therapeutic hypothermia for cardiac arrest in adults and hypoxic-ischemic injury in infants. Similarly, clinical interest in hypothermia for other brain injuries, such as stroke, has been greatly supported by positive findings in preclinical work. The reliability, validity, and utility of animal models, among many research practices (blinding, randomization, etc.), are key to successful clinical translation. Here, we review methods used to induce and maintain hypothermia in animal models. These include physical and pharmacological methods. We emphasize the advantages and limitations of each approach, and the importance of using clinically relevant cooling protocols and appropriate monitoring and reporting approaches. Moreover, we performed a literature survey of ischemic stroke studies published in 2015 to highlight the continuing risk of temperature confounds in neuroprotection studies. For example, many still do not accurately monitor and report temperature during surgery (23.5%), even though almost half of these studies (46.0%) use pharmaceutical agents that likely influence temperature. We hope this review stimulates awareness and discussion of the importance of temperature in neuroprotective studies.
Collapse
Affiliation(s)
- Ana C Klahr
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Canada
| | - Colby A Nadeau
- 2 Department of Psychology, University of Alberta , Edmonton, Canada
| | - Frederick Colbourne
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Canada .,2 Department of Psychology, University of Alberta , Edmonton, Canada
| |
Collapse
|
27
|
Abstract
About half of patients survive intracerebral hemorrhage (ICH), but most are left with significant disability. Rehabilitation after ICH is the mainstay of treatment to reduce impairment, improve independence in activities, and return patients to meaningful participation in the community. The authors discuss the neuroplastic mechanisms underlying recovery in ICH, preclinical and clinical interventional studies to augment recovery, and the rehabilitative and medical management of post-ICH patients.
Collapse
Affiliation(s)
- Michael F Saulle
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York
| | - Heidi M Schambra
- Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, New York
| |
Collapse
|
28
|
Wu CH, Chen CC, Lai CY, Hung TH, Lin CC, Chao M, Chen SF. Treatment with TO901317, a synthetic liver X receptor agonist, reduces brain damage and attenuates neuroinflammation in experimental intracerebral hemorrhage. J Neuroinflammation 2016; 13:62. [PMID: 26968836 PMCID: PMC4788882 DOI: 10.1186/s12974-016-0524-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/03/2016] [Indexed: 11/10/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) induces a series of inflammatory processes that contribute to neuronal damage and neurological deterioration. Liver X receptors (LXRs) are nuclear receptors that negatively regulate transcriptional processes involved in inflammatory responses, but their role in the pathology following ICH remains unclear. The present study investigated the neuroprotective effects and anti-inflammatory actions of TO901317, a synthetic LXR agonist, in a model of collagenase-induced ICH and in microglial cultures. Methods Mice subjected to collagenase-induced ICH injury were injected with either TO901317 (30 mg/kg) or vehicle 10 min after ICH and subsequently daily for 2 days. Behavioral studies, histology analysis, and assessments of hematoma volumes, brain water content, and blood-brain barrier (BBB) permeability were performed. The protein expression of LXR-α, LXR-β, ATP binding cassette transporter-1 (ABCA-1), and inflammatory molecules was analyzed. The anti-inflammatory mechanism of TO901317 was investigated in cultured microglia that were stimulated with either lipopolysaccharide (LPS) or thrombin. Results ICH induced an increase in LXR-α protein levels in the hemorrhagic hemisphere at 6 h whereas LXR-β expression remained unaffected. Both LXR-α and LXR-β were expressed in neurons and microglia in the peri-ICH region and but rarely in astrocytes. TO901317 significantly attenuated functional deficits and brain damage up to 28 days post-ICH. TO901317 also reduced neuronal death, BBB disruption, and brain edema at day 4 post-ICH. These changes were associated with marked reductions in microglial activation, neutrophil infiltration, and expression levels of inflammatory mediators at 4 and 7 days. However, TO901317 had no effect on matrix metalloproteinase-9 activity. In BV2 microglial cultures, TO901317 attenuated LPS- and thrombin-stimulated nitric oxide production and reduced LPS-induced p38, JNK, MAPK, and nuclear factor-kappa B (NF-κB) signaling. Moreover, delaying administration of TO901317 to 3 h post-ICH reduced brain tissue damage and neuronal death. Conclusions Our results suggest that enhancing LXR activation may provide a potential therapy for ICH by modulating the cytotoxic functions of microglia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0524-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chun-Hu Wu
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chai-You Lai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Taipei and College of Medicine, Chang Gung University, Taipei, Taiwan, Republic of China
| | - Chao-Chang Lin
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Min Chao
- School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China.
| |
Collapse
|
29
|
After Intracerebral Hemorrhage, Oligodendrocyte Precursors Proliferate and Differentiate Inside White-Matter Tracts in the Rat Striatum. Transl Stroke Res 2016; 7:192-208. [PMID: 26743212 PMCID: PMC4873533 DOI: 10.1007/s12975-015-0445-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/17/2015] [Accepted: 12/28/2015] [Indexed: 02/06/2023]
Abstract
Damage to myelinated axons contributes to neurological deficits after acute CNS injury, including ischemic and hemorrhagic stroke. Potential treatments to promote re-myelination will require fully differentiated oligodendrocytes, but almost nothing is known about their fate following intracerebral hemorrhage (ICH). Using a rat model of ICH in the striatum, we quantified survival, proliferation, and differentiation of oligodendrocyte precursor cells (OPCs) (at 1, 3, 7, 14, and 28 days) in the peri-hematoma region, surrounding striatum, and contralateral striatum. In the peri-hematoma, the density of Olig2+ cells increased dramatically over the first 7 days, and this coincided with disorganization and fragmentation of myelinated axon bundles. Very little proliferation (Ki67+) of Olig2+ cells was seen in the anterior subventricular zone from 1 to 28 days. However, by 3 days, many were proliferating in the peri-hematoma region, suggesting that local proliferation expands their population. By 14 days, the density of Olig2+ cells declined in the peri-hematoma region, and, by 28 days, it reached the low level seen in the contralateral striatum. At these later times, many surviving axons were aligned into white-matter bundles, which appeared less swollen or fragmented. Oligodendrocyte cell maturation was prevalent over the 28-day period. Densities of immature OPCs (NG2+Olig2+) and mature (CC-1+Olig2+) oligodendrocytes in the peri-hematoma increased dramatically over the first week. Regardless of the maturation state, they increased preferentially inside the white-matter bundles. These results provide evidence that endogenous oligodendrocyte precursors proliferate and differentiate in the peri-hematoma region and have the potential to re-myelinate axon tracts after hemorrhagic stroke.
Collapse
|
30
|
Mechanisms of Cerebral Hemorrhage. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Klahr AC, Dietrich K, Dickson CT, Colbourne F. Prolonged Localized Mild Hypothermia Does Not Affect Seizure Activity After Intracerebral Hemorrhage in Rats. Ther Hypothermia Temp Manag 2015; 6:40-7. [PMID: 26717112 DOI: 10.1089/ther.2015.0028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating stroke with high morbidity and mortality. Post-ICH seizures are a common complication, potentially increasing brain injury and the risk of delayed epilepsy. Mild therapeutic hypothermia (HYPO, ∼33°C) is neuroprotective against several brain insults, such as ischemia, while also mitigating seizure incidence and severity in some instances. Therefore, we tested whether brain-selective HYPO reduced electrographic seizure activity after a collagenase-induced striatal ICH in rats. Animals were injected unilaterally with 0.14 U of bacterial collagenase, implanted with a unilateral brain cooling device, and a probe to bilaterally record electroencephalographic (EEG) activity. Cooling began 6 hours after collagenase infusion and was maintained for 48 hours, followed by rewarming over 6 hours. Our protocol did not affect EEG activity in naïve rats, nor did it increase bleeding after ICH (∼50 μL). Epileptiform activity commonly occurred in untreated ICH rats (∼60% of animals), but HYPO did not affect the incidence, timing, total duration of seizures, or the peaks occurring during epileptiform activity. However, longer average duration was detected on the ipsilateral side to stroke in the HYPO group (p < 0.05). Cooling did not affect neurological deficits (e.g., circling), measured 7 and 14 days after ICH, or lesion volume (∼35 mm(3)). In addition, there was no relationship among endpoints (e.g., seizures and lesion size). In summary, HYPO failed to reduce seizure activity after ICH, which fits with our separate findings that cooling does not mitigate thrombin and iron-mediated secondary injury mechanisms thought to cause seizures after ICH. Additional research is needed to identify better HYPO protocols and the use of cotreatments to maximize the benefit of HYPO to ICH patients.
Collapse
Affiliation(s)
- Ana C Klahr
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Canada
| | - Kristen Dietrich
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Canada
| | - Clayton T Dickson
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Canada .,2 Department of Psychology, University of Alberta , Edmonton, Canada
| | - Frederick Colbourne
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Canada .,2 Department of Psychology, University of Alberta , Edmonton, Canada
| |
Collapse
|
32
|
John RF, Colbourne F. Delayed localized hypothermia reduces intracranial pressure following collagenase-induced intracerebral hemorrhage in rat. Brain Res 2015; 1633:27-36. [PMID: 26723566 DOI: 10.1016/j.brainres.2015.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 01/30/2023]
Abstract
Brain injury, such as from intracerebral hemorrhage (ICH), causes edema and raises intracranial pressure (ICP)--a potentially life-threatening complication. Clinical studies suggest that therapeutic hypothermia (TH) reduces edema and ICP after ICH. Similarly, animal studies show that TH can sometimes reduce edema, but whether ICP would be attenuated is not known. Here we tested whether 24-h delayed TH reduces edema and ICP in rats with severe striatal ICH (collagenase model). First, we showed that ICH increased epidural ICP (mean of 18 vs. 6.5mm Hg in controls), measured via telemetry. Second, we confirmed that delayed TH did not affect hematoma size at 7d ay (~65 vs. ~61 µL in controls). A cranial cooling device lowered striatal temperature to ~33 °C from 24 to 72 h after ICH. Third, we compared normothermic rats to those with TH that were rewarmed immediately or over 6h. Both TH protocols significantly reduced average and peak ICP by the second treatment day, and benefits persisted after rewarming. However, TH with slow rewarming failed to mitigate edema at 96 h (83.2% vs. 83.6% in controls) whereas rapid rewarming worsened edema (85.7%). Finally, we compared normothermic and TH rats without rewarming and found no impact on edema at 72 h (~81%). In summary, it appears that 24-h delayed local TH lowers ICP by a mechanism other than edema. Rapid rewarming worsens edema after local cooling, but this did not markedly impact ICP. Thus, TH should reduce ICP in patients with severe ICH, but not necessarily through mitigating edema.
Collapse
Affiliation(s)
- Roseleen F John
- Neuroscience and Mental Health Institute University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute University of Alberta, Edmonton, Alberta, Canada; Department of Psychology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
33
|
Semyachkina-Glushkovskaya O, Pavlov A, Kurths J, Borisova E, Gisbrecht A, Sindeeva O, Abdurashitov A, Shirokov A, Navolokin N, Zinchenko E, Gekalyuk A, Ulanova M, Zhu D, Luo Q, Tuchin V. Optical monitoring of stress-related changes in the brain tissues and vessels associated with hemorrhagic stroke in newborn rats. BIOMEDICAL OPTICS EXPRESS 2015; 6:4088-97. [PMID: 26504656 PMCID: PMC4605065 DOI: 10.1364/boe.6.004088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/26/2015] [Accepted: 09/18/2015] [Indexed: 05/04/2023]
Abstract
Stress is a major factor for a risk of cerebrovascular catastrophes. Studying of mechanisms underlying stress-related brain-injures in neonates is crucial for development of strategy to prevent of neonatal stroke. Here, using a model of sound-stress-induced intracranial hemorrhages in newborn rats and optical methods, we found that cerebral veins are more sensitive to the deleterious effect of stress than arteries and microvessels. The development of venous insufficiency with decreased blood outflow from the brain accompanied by hypoxia, reduction of complexity of venous blood flow and high production of beta-arrestin-1 are possible mechanisms responsible for a risk of neonatal hemorrhagic stroke.
Collapse
Affiliation(s)
- Oxana Semyachkina-Glushkovskaya
- Saratov State University, Astrakhanskaya Str. 83, Saratov 410012, Russia
- Huazhong University of Science and Technology, Wuhan 430074, China
| | - Alexey Pavlov
- Saratov State University, Astrakhanskaya Str. 83, Saratov 410012, Russia
| | - Jürgen Kurths
- Huazhong University of Science and Technology, Wuhan 430074, China
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473 Potsdam, Germany
| | - Ekaterina Borisova
- Institute of Electronics, Bulgarian Academy of Sciences, Tsarigradsko Chaussee 72, Sofia 1784, Bulgaria
| | - Alexander Gisbrecht
- Institute of Electronics, Bulgarian Academy of Sciences, Tsarigradsko Chaussee 72, Sofia 1784, Bulgaria
| | - Olga Sindeeva
- Saratov State University, Astrakhanskaya Str. 83, Saratov 410012, Russia
| | | | - Alexander Shirokov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Entusiastov Str.13, Saratov 410049, Russia
| | | | | | - Artem Gekalyuk
- Saratov State University, Astrakhanskaya Str. 83, Saratov 410012, Russia
| | - Maria Ulanova
- Saratov State University, Astrakhanskaya Str. 83, Saratov 410012, Russia
| | - Dan Zhu
- Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingming Luo
- Huazhong University of Science and Technology, Wuhan 430074, China
| | - Valery Tuchin
- Saratov State University, Astrakhanskaya Str. 83, Saratov 410012, Russia
- Huazhong University of Science and Technology, Wuhan 430074, China
- Laboratory of Biophotonics, Tomsk State University, Tomsk 634050, Russia
| |
Collapse
|
34
|
Thrombin-induced microglial activation impairs hippocampal neurogenesis and spatial memory ability in mice. Behav Brain Funct 2015; 11:30. [PMID: 26410080 PMCID: PMC4584127 DOI: 10.1186/s12993-015-0075-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 09/11/2015] [Indexed: 11/10/2022] Open
Abstract
Background To investigate the effects of microglia/macrophages activation induced by intrastriatal thrombin injection on dentate gyrus neurogenesis and spatial memory ability in mice. Methods The male C57BL/6 mice were divided into 4 groups of 10: sham, intracerebral hemorrhage (ICH), ICH + hirudin (thrombin inhibitor), and ICH + indometacin (Indo, an anti-inflammation drug). ICH model was created by intrastriatal thrombin (1U) injection. BrdU (50 mg/kg) was administrated on the same day after surgery for 6 consecutive days. Motor functions were evaluated with rotarod and beam walking tests. The spatial memory deficit was measured with Morris water maze (MWM). Cell quantification was performed for doublecortin (DCX, immature neuron), BrdU (S-phase proliferating cell population) and CD68 (activated microglia/macrophage) immune-reactive cells. Results Microglia/macrophages activation induced by intrastriatal thrombin injection reduced hippocampal neurogenesis and impaired spatial memory ability, but did not affect the motor function at 3 and 5 days post-injury. Both hirudin and indometacin reduced microglia/macrophages activation, enhanced hippocampal neurogenesis, and improved spatial memory ability in mice. Conclusions Microglia/macrophages activation induced by intrastriatal thrombin injection might be responsible for the spatial memory deficit. Targeting both thrombin and inflammation systems in acute phase of ICH might be important in alleviating the significant spatial memory deficits.
Collapse
|
35
|
Hackett MJ, DeSouza M, Caine S, Bewer B, Nichol H, Paterson PG, Colbourne F. A new method to image heme-Fe, total Fe, and aggregated protein levels after intracerebral hemorrhage. ACS Chem Neurosci 2015; 6:761-70. [PMID: 25695130 DOI: 10.1021/acschemneuro.5b00037] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
An intracerebral hemorrhage (ICH) is a devastating stroke that results in high mortality and significant disability in survivors. Unfortunately, the underlying mechanisms of this injury are not yet fully understood. After the primary (mechanical) trauma, secondary degenerative events contribute to ongoing cell death in the peri-hematoma region. Oxidative stress is thought to be a key reason for this delayed injury, which is likely due to free-Fe-catalyzed free radical reactions. Unfortunately, this is difficult to prove with conventional biochemical assays that fail to differentiate between alterations that occur within the hematoma and peri-hematoma zone. This is a critical limitation, as the hematoma contains tissue severely damaged by the initial hemorrhage and is unsalvageable, whereas the peri-hematoma region is less damaged but at risk from secondary degenerative events. Such events include oxidative stress mediated by free Fe presumed to originate from hemoglobin breakdown. Therefore, minimizing the damage caused by oxidative stress following hemoglobin breakdown and Fe release is a major therapeutic target. However, the extent to which free Fe contributes to the pathogenesis of ICH remains unknown. This investigation used a novel imaging approach that employed resonance Raman spectroscopic mapping of hemoglobin, X-ray fluorescence microscopic mapping of total Fe, and Fourier transform infrared spectroscopic imaging of aggregated protein following ICH in rats. This multimodal spectroscopic approach was used to accurately define the hematoma/peri-hematoma boundary and quantify the Fe concentration and the relative aggregated protein content, as a marker of oxidative stress, within each region. The results revealed total Fe is substantially increased in the hematoma (0.90 μg cm(-2)), and a subtle but significant increase in Fe that is not in the chemical form of hemoglobin is present within the peri-hematoma zone (0.32 μg cm(-2)) within 1 day of ICH, relative to sham animals (0.22 μg cm(-2)). Levels of aggregated protein were significantly increased within both the hematoma (integrated band area 0.10 AU) and peri-hematoma zone (integrated band area 0.10 AU) relative to sham animals (integrated band area 0.056 AU), but no significant difference in aggregated protein content was observed between the hematoma and peri-hematoma zone. This result suggests that the chemical form of Fe and its ability to generate free radicals is likely to be a more critical predictor of tissue damage than the total Fe content of the tissue. Furthermore, this article describes a novel approach to colocalize nonheme Fe and aggregated protein in the peri-hematoma zone following ICH, a significant methodological advancement for the field.
Collapse
Affiliation(s)
- Mark J. Hackett
- Molecular
and Environmental Sciences Group, Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Mauren DeSouza
- Department
of Psychology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
- Stress,
Memory and Behaviour Lab, Graduate Program in Biochemistry, Federal University of Pampa, Uruguaiana, Rio Grande do Sul 97500-970, Brazil
| | - Sally Caine
- Department
of Anatomy and Cell Biology, University of Saskatchewan, 107
Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Brian Bewer
- Canadian Light Source, Saskatoon, Saskatchewan S7N 2V3, Canada
| | - Helen Nichol
- Department
of Anatomy and Cell Biology, University of Saskatchewan, 107
Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Phyllis G. Paterson
- College of
Pharmacy and Nutrition, University of Saskatchewan, D Wing Health Sciences, 107 Wiggins
Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Frederick Colbourne
- Department
of Psychology and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| |
Collapse
|
36
|
Stem cell-based therapies for intracerebral hemorrhage in animal model: a meta-analysis. Neurol Sci 2015; 36:1311-7. [PMID: 25972140 DOI: 10.1007/s10072-015-2238-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/09/2015] [Indexed: 11/27/2022]
Abstract
Stem cell to be a new intervention for treating intracerebral hemorrhage (ICH) might benefit humans. Therefore, we collected animal studies to find the effect of this innovative treatment. In July 2014, we searched Medline (from 1950), Embase (from 1980), China Biology Medicine disk (from 1978) for studies on stem cells used for treating experimental ICH in animal models that reported neurobehavioral and structural outcome. We evaluated the quality of these studies and used a weighted mean difference random affects model for the meta-analysis. We have collected 30 studies from 650 publications identified through systematic review describing the effects of 5 different type of stem cells on 12 different neurobehavioral scales with 1101 rodents or monkeys. Although there is lack of uniformity of the evaluation methods, these researches showed consistent improvements both in neurobehavioral function and structural outcomes. Besides, the quality of these studies needs to be raised. In conclusion, stem cells hold extensive potential in treating ICH, which should be further evaluated with more evidence-based, high-quality animal studies.
Collapse
|
37
|
Chang CF, Cai L, Wang J. Translational intracerebral hemorrhage: a need for transparent descriptions of fresh tissue sampling and preclinical model quality. Transl Stroke Res 2015; 6:384-9. [PMID: 25907620 DOI: 10.1007/s12975-015-0399-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/19/2022]
Abstract
For years, strategies have been proposed to improve translational success in stroke research by improving the quality of animal studies. However, articles that report preclinical intracerebral hemorrhage (ICH) studies continue to lack adequate qualitative and quantitative descriptions of fresh brain tissue collection. They also tend to lack transparency about animal model quality. We conducted a systematic review of 82 ICH research articles to determine the level of detail reported for brain tissue collection. We found that only 24 (29 %) reported the volume, weight, or thickness of tissue collected and a specific description of the anatomical location. Thus, up to 71 % of preclinical ICH research articles did not properly define how fresh specimens were collected for biochemical measurements. Such omissions may impede reproducibility of results between laboratories. Although existing criteria have improved the quality of preclinical stroke studies, ICH researchers need to identify specific guidelines and strategies to avoid pitfalls, minimize bias, and increase reproducibility in this field.
Collapse
Affiliation(s)
- Che-Feng Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross Bldg 370B, Baltimore, MD, 21205, USA
| | | | | |
Collapse
|
38
|
Höllig A, Weinandy A, Nolte K, Clusmann H, Rossaint R, Coburn M. Experimental subarachnoid hemorrhage in rats: comparison of two endovascular perforation techniques with respect to success rate, confounding pathologies and early hippocampal tissue lesion pattern. PLoS One 2015; 10:e0123398. [PMID: 25867893 PMCID: PMC4395040 DOI: 10.1371/journal.pone.0123398] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/18/2015] [Indexed: 11/30/2022] Open
Abstract
Recently aside from the “classic” endovascular monofilament perforation technique to induce experimental subarachnoid hemorrhage (SAH) a modification using a tungsten wire advanced through a guide tube has been described. We aim to assess both techniques for their success rate (induction of SAH without confounding pathologies) as primary endpoint. Further, the early tissue lesion pattern as evidence for early brain injury will be analyzed as secondary endpoint. Sprague Dawley rats (n=39) were randomly assigned to receive either Sham surgery (n=4), SAH using the “classic” technique (n=18) or using a modified technique (n=17). Course of intracranial pressure (ICP) and regional cerebral blood flow (rCBF) was analyzed; subsequent pathologies were documented either 6 or 24 h after SAH. Hippocampal tissue samples were analyzed via immunohistochemistry and western blotting. SAH-induction, regardless of confounding pathologies, was independent from type of technique (p=0.679). There was no significant difference concerning case fatality rate (classic: 40%; modified: 20%; p=0.213). Successful induction of SAH without collateral ICH or SDH was possible in 40% with the classic and in 86.7% with the modified technique (p=0.008). Peak ICP levels differed significantly between the two groups (classic: 94 +/- 23 mmHg; modified: 68 +/- 19 mmHg; p=0.003). Evidence of early cellular stress response and activation of apoptotic pathways 6 h after SAH was demonstrated. The extent of stress response is not dependent on type of technique. Both tested techniques successfully produce SAH including activation of an early stress response and apoptotic pathways in the hippocampal tissue. However, the induction of SAH with less confounding pathologies was more frequently achieved with the modified tungsten wire technique.
Collapse
Affiliation(s)
- Anke Höllig
- Department of Neurosurgery, University RWTH Aachen, Aachen, Germany
- Department of Anesthesiology, University RWTH Aachen, Aachen, Germany
| | - Agnieszka Weinandy
- Department of Neurosurgery, University RWTH Aachen, Aachen, Germany
- Department of Neuropathology, University RWTH Aachen, Aachen, Germany
| | - Kay Nolte
- Department of Neuropathology, University RWTH Aachen, Aachen, Germany
| | - Hans Clusmann
- Department of Neurosurgery, University RWTH Aachen, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, University RWTH Aachen, Aachen, Germany
| | - Mark Coburn
- Department of Anesthesiology, University RWTH Aachen, Aachen, Germany
- * E-mail:
| |
Collapse
|
39
|
Vieira de Castro AC, Olsson IAS. Does the goal justify the methods? Harm and benefit in neuroscience research using animals. Curr Top Behav Neurosci 2015; 19:47-78. [PMID: 24844681 DOI: 10.1007/7854_2014_319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The goal of the present chapter is to open up for discussion some of the major ethical issues involved in animal-based neuroscience research. We begin by approaching the question of the moral acceptability of the use of animals in research at all, exploring the implications of three different ethical theories: contractarianism, utilitarianism, and animal rights. In the rest of this chapter, we discuss more specific issues of neuroscience research within what we argue is the mainstream framework for research animal ethics, namely one based on harm-benefit analysis. We explore issues of harms and benefits and how to balance them as well as how to reduce harm and increase benefit within neuroscience research.
Collapse
Affiliation(s)
- Ana Catarina Vieira de Castro
- Laboratory Animal Science Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | | |
Collapse
|
40
|
Wowk S, Ma Y, Colbourne F. Mild Therapeutic Hypothermia Does Not Reduce Thrombin-Induced Brain Injury. Ther Hypothermia Temp Manag 2014; 4:180-7. [DOI: 10.1089/ther.2014.0014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shannon Wowk
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Yonglie Ma
- Department of Psychology, University of Alberta, Edmonton, Canada
| | - Frederick Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Psychology, University of Alberta, Edmonton, Canada
| |
Collapse
|
41
|
John RF, Williamson MR, Dietrich K, Colbourne F. Localized hypothermia aggravates bleeding in the collagenase model of intracerebral hemorrhage. Ther Hypothermia Temp Manag 2014; 5:19-25. [PMID: 25386695 DOI: 10.1089/ther.2014.0020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Animal studies testing whether therapeutic hypothermia is neuroprotective after intracerebral hemorrhage (ICH) have been inconclusive. In rodents, ICH is often produced in the striatum by infusing collagenase, which causes prolonged hemorrhaging from multiple vessels. Our previous data shows that this bleeding (hematoma) is worsened by systemic hypothermia given soon after collagenase infusion. In this study we hypothesized that localized brain hypothermia would also aggravate bleeding in this model (0.2 U of collagenase in 1.2 μL of saline). We also evaluated cooling after intrastriatal thrombin infusion (1 U in 30 μL of saline)-a simplified model of ICH thought to cause bleeding. Focal hypothermia was achieved by flushing cold water through an implanted cooling device attached to the skull underneath the temporalis muscle of adult rats. Previous work and data at this time shows this method cools the striatum to ∼33°C, whereas the body remains normothermic. In comparison to normothermic groups, cooling significantly worsened bleeding when instituted at 6 hours (∼94 vs. 42 μL, p=0.018) and 12 hours (79 vs. 61 μL, p=0.042) post-ICH (24-hour survival), but not after a 24-hour delay (36-hour survival). Rats were cooled until euthanasia when hematoma size was determined by a hemoglobin-based spectrophotometry assay. Cooling did not influence cerebral blood volume after just saline or thrombin infusion. The latter is explained by the fact that thrombin did not cause bleeding beyond that caused by saline infusion. In summary, local hypothermia significantly aggravates bleeding many hours after collagenase infusion suggesting that bleeding may have confounded earlier studies with hypothermia. Furthermore, these findings serve as a cautionary note on using cooling even many hours after cerebral bleeding.
Collapse
Affiliation(s)
- Roseleen F John
- 1 Neuroscience and Mental Health Institute, University of Alberta , Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
42
|
Abstract
BACKGROUND Brain injury after intracerebral hemorrhage (ICH) arises from numerous contributors, of which some also play essential roles. Notably, thrombin production, needed to stop bleeding, also causes acute cell death and edema. In some rodent models of ICH, peri-hematoma neurons die over weeks. Hence we evaluated whether thrombin is responsible for this chronic degeneration. Functional impairments after ICH also result from sub-lethal damage to neurons, especially the loss of dendrites. Thus, we evaluated whether thrombin infusion alone, a reductionist model of ICH, causes similar injury. METHODS Adult rats had a modest intra-striatal infusion of thrombin (1 U) or saline followed by a behavioral test, to verify impairment, 7 days later. After this they were euthanized and tissue stained with Golgi-Cox solution to allow the assessment of dendritic morphology in striatal neurons. In a second experiment, rats survived 7 or 60 days after thrombin infusion in order to histologically determine lesion volume. RESULTS Thrombin caused early cell death and considerable atrophy in surviving peri-lesion neurons, which had less than half of their usual numbers of branches. However, total tissue loss was comparable at 7 (24.1 mm3) and 60 days (25.6 mm3). CONCLUSION Thrombin infusion causes early cell death and neuronal atrophy in nearby surviving striatal neurons but thrombin does not cause chronic tissue loss. Thus, the chronic degeneration found after ICH in rats is not simply and solely due to acute thrombin production. Nonetheless, thrombin is an important contributor to behavioral dysfunction because it causes cell death and substantial dendritic injury.
Collapse
|
43
|
Seizure activity occurs in the collagenase but not the blood infusion model of striatal hemorrhagic stroke in rats. Transl Stroke Res 2014; 6:29-38. [PMID: 25053257 PMCID: PMC4297611 DOI: 10.1007/s12975-014-0361-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/04/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
Abstract
Seizures are a frequent complication of brain injury, including intracerebral hemorrhage (ICH), where seizures occur in about a third of patients. Rodents are used to study pathophysiology and neuroprotective therapies after ICH, but there have been no studies assessing the occurrence of seizures in these models. Thus, we compared seizure incidence and characteristics after infusing collagenase (0.14 U), which degrades blood vessels, and autologous blood (100 μL) into the striatum of rats. Saline was infused in others as a negative control, whereas iron, a by-product of degrading erythrocytes, served as a positive control. Ipsilateral and contralateral electroencephalographic (EEG) activity was continuously monitored with telemetry probes for a week after the stroke. There were no electrographic abnormalities during baseline recordings. As expected, saline did not elicit any epileptiform activity whereas iron caused seizure activity. Seizures occurred in 66 % of the collagenase group between 10 and 36 h, their duration ranged from 5 to 90 s, and these events were mostly observed bilaterally. No such activity occurred after blood infusion despite comparable lesion sizes of 32.5 and 40.9 mm3 in the collagenase and blood models, respectively (p = 0.222). Therefore, seizures are a common acute occurrence in the collagenase but not whole blood models of striatal ICH (p = 0.028, for incidence). These findings have potential implications for ICH studies such as for understanding model differences, helping select which model to use, and determining how seizures may affect or be affected by treatments applied after stroke.
Collapse
|
44
|
Zhu W, Gao Y, Chang CF, Wan JR, Zhu SS, Wang J. Mouse models of intracerebral hemorrhage in ventricle, cortex, and hippocampus by injections of autologous blood or collagenase. PLoS One 2014; 9:e97423. [PMID: 24831292 PMCID: PMC4022524 DOI: 10.1371/journal.pone.0097423] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 04/17/2014] [Indexed: 01/08/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition. Existing preclinical ICH models focus largely on striatum but neglect other brain areas such as ventricle, cortex, and hippocampus. Clinically, however, hemorrhagic strokes do occur in these other brain regions. In this study, we established mouse hemorrhagic models that utilize stereotactic injections of autologous whole blood or collagenase to produce ventricular, cortical, and hippocampal injury. We validated and characterized these models by histology, immunohistochemistry, and neurobehavioral tests. In the intraventricular hemorrhage (IVH) model, C57BL/6 mice that received unilateral ventricular injections of whole blood demonstrated bilateral ventricular hematomas, ventricular enlargement, and brain edema in the ipsilateral cortex and basal ganglia at 72 h. Unilateral injections of collagenase (150 U/ml) caused reproducible hematomas and brain edema in the frontal cortex in the cortical ICH (c-ICH) model and in the hippocampus in the hippocampal ICH (h-ICH) model. Immunostaining revealed cellular inflammation and neuronal death in the periventricular regions in the IVH brain and in the perihematomal regions in the c-ICH and h-ICH brains. Locomotor abnormalities measured with a 24-point scoring system were present in all three models, especially on days 1, 3, and 7 post-ICH. Locomotor deficits measured by the wire-hanging test were present in models of IVH and c-ICH, but not h-ICH. Interestingly, mice in the c-ICH model demonstrated emotional abnormality, as measured by the tail suspension test and forced swim test, whereas h-ICH mice exhibited memory abnormality, as measured by the novel object recognition test. All three ICH models generated reproducible brain damage, brain edema, inflammation, and consistent locomotor deficits. Additionally, the c-ICH model produced emotional deficits and the h-ICH model produced cognitive deficits. These three models closely mimic human ICH and should be useful for investigating the pathophysiology of ICH in ventricle, cortex, and hippocampus and for evaluating potential therapeutic strategies.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Yufeng Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Che-Feng Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Jie-ru Wan
- Department of Biological Sciences, Illinois Institute of Technology, College of Science, Chicago, Illinois, United States of America
| | - Shan-shan Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
45
|
Chang CF, Cho S, Wang J. (-)-Epicatechin protects hemorrhagic brain via synergistic Nrf2 pathways. Ann Clin Transl Neurol 2014; 1:258-271. [PMID: 24741667 PMCID: PMC3984761 DOI: 10.1002/acn3.54] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective In the wake of intracerebral hemorrhage (ICH), a devastating stroke with no effective treatment, hemoglobin/iron-induced oxidative injury leads to neuronal loss and poor neurologic outcomes. (-)-Epicatechin (EC), a brain-permeable flavanol that modulates redox/oxidative stress via the NF-E2–related factor (Nrf) 2 pathway, has been shown to be beneficial for vascular and cognitive function in humans. Here, we examined whether EC can reduce early brain injury in ICH mouse models and investigated the underlying mechanisms. Methods ICH was induced by injecting collagenase, autologous blood, or thrombin into mouse striatum. EC was administered orally at 3 h after ICH and then every 24 h. Lesion volume, neurologic deficits, brain edema, reactive oxygen species, and protein expression and activity were evaluated. Results EC significantly reduced lesion volume and ameliorated neurologic deficits in both male and female ICH mice. Cell death and neuronal degeneration were decreased in the perihematomal area and were associated with reductions in caspase-3 activity and high-mobility group protein B1 (HMGB-1) level. These changes were accompanied by attenuation of oxidative insults, increased phase II enzyme expression, and increased Nrf2 nuclear accumulation. Interestingly, in addition to providing neuroprotection via Nrf2 signaling, EC diminished heme oxygenase-1 induction and brain iron deposition via an Nrf2-independent pathway that downregulated ICH-induced activating protein-1 activation and decreased matrix metalloproteinase 9 activity, lipocalin-2 levels, iron-dependent cell death, and ferroptosis-related gene expression. Interpretation Collectively, our data show that EC protects against ICH by activation of Nrf2-dependent and -independent pathways and may serve as a potential intervention for patients with ICH.
Collapse
Affiliation(s)
- Che-Feng Chang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Suzy Cho
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
46
|
Hiploylee C, Colbourne F. Intracranial pressure measured in freely moving rats for days after intracerebral hemorrhage. Exp Neurol 2014; 255:49-55. [PMID: 24582611 DOI: 10.1016/j.expneurol.2014.02.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 02/09/2014] [Accepted: 02/20/2014] [Indexed: 10/25/2022]
Abstract
In some patients, intracerebral hemorrhage (ICH) causes life-threatening elevations in intracranial pressure (ICP) arising from mass effect of the hematoma and edema. Accordingly, edema is a common endpoint to gauge treatment efficacy in rodent ICH models. Despite widespread reliance on edema, its relationship with ICP and cerebral perfusion pressure (CPP) is unknown. Blood pressure (BP) and ICP were measured by telemetry devices in rats after collagenase ("severe" - 0.3U, and "moderate" - 0.15U doses) or blood infusion (100μL) into striatum (vs. saline infused shams). We compared epidural and intraparenchymal ICP readings (collagenase), evaluated CPP (collagenase), and compared models. Moderate (9.46mmHg±4.72 SD, 3day average) and severe collagenase ICHs (10.79±3.50) significantly increased ICP versus shams (4.02±2.09), whereas blood infusion did not (5.37±0.55). The two monitoring locations gave similar readings after severe collagenase ICH. Increased ICP reduced CPP by ~7.5mmHg for days after the larger collagenase infusion. CPP averaged from 103-112mmHg in shams. Edema occurred in all ICH models and predicted ICP. However, ICP and CPP were only modestly changed even after severe ICH and edema. Thus, small changes in edema typically reported in the literature, which often use smaller bleeds than presently used, likely minimally affects ICP and CPP. Further research into the face validity of these models, endpoints, and their ability to evaluate therapeutics is needed.
Collapse
Affiliation(s)
- Carmen Hiploylee
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada
| | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta, Canada; Center for Neuroscience, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
47
|
Caliaperumal J, Colbourne F. Rehabilitation Improves Behavioral Recovery and Lessens Cell Death Without Affecting Iron, Ferritin, Transferrin, or Inflammation After Intracerebral Hemorrhage in Rats. Neurorehabil Neural Repair 2013; 28:395-404. [DOI: 10.1177/1545968313517758] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background. Rehabilitation aids recovery from stroke in animal models, including in intracerebral hemorrhage (ICH). Sometimes, rehabilitation lessens brain damage. Objective. We tested whether rehabilitation improves recovery and reduces perihematoma neuronal death. We also evaluated whether rehabilitation influences iron toxicity and inflammation, mediators of secondary degeneration after ICH. Methods. Rats were trained to retrieve food pellets in a staircase apparatus and later subjected to striatal ICH (via collagenase infusion). After 1 week, they were given either enriched rehabilitation (ER), including reach training with group housing and environmental enrichment, or control treatment (group housing). Rats in the first experiment were treated for 2 weeks, functionally assessed, and killed humanely at 1 month to determine brain levels of nonheme iron. A second experiment used a similar approach, except that animals were euthanized at 14 days to evaluate perihematoma neuronal death (FluoroJade), iron distribution (Perls), and astrocyte (GFAP) and microglia (Iba-1) activity. A third experiment measured levels of iron-binding proteins (ferritin and transferrin) at 14 days. Results. Striatal ICH caused functional impairments, which were significantly improved with ER. The ICH caused delayed perihematoma neuronal death, which ER significantly reduced. Hemispheric iron levels, the amount of iron-binding proteins, and perihematoma astrocytes and microglia numbers were significantly elevated after ICH (vs normal side) but were not affected by ER. Conclusions. Rehabilitation is an effective behavioral and neuroprotective strategy for ICH. Neither effect appears to stem from influencing iron toxicity or inflammation. Thus, additional work must identify underlying mechanisms to help further therapeutic gains.
Collapse
|
48
|
Bipyridine, an iron chelator, does not lessen intracerebral iron-induced damage or improve outcome after intracerebral hemorrhagic stroke in rats. Transl Stroke Res 2013; 4:719-28. [PMID: 24323426 DOI: 10.1007/s12975-013-0272-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 07/18/2013] [Accepted: 07/19/2013] [Indexed: 10/26/2022]
Abstract
Iron chelators, such as the intracellular ferrous chelator 2,2'-bipyridine, are a potential means of ameliorating iron-induced injury after intracerebral hemorrhage (ICH). We evaluated bipyridine against the collagenase and whole-blood ICH models and a simplified model of iron-induced damage involving a striatal injection of FeCl2 in adult rats. First, we assessed whether bipyridine (25 mg/kg beginning 12 h post-ICH and every 12 h for 3 days) would attenuate non-heme iron levels in the brain and lessen behavioral impairments (neurological deficit scale, corner turn test, and horizontal ladder) 7 days after collagenase-induced ICH. Second, we evaluated bipyridine (20 mg/kg beginning 6 h post-ICH and then every 24 h) on edema 3 days after collagenase infusion. Body temperature was continually recorded in a subset of these rats beginning 24 h prior to ICH until euthanasia. Third, bipyridine was administered (as per experiment 2) after whole-blood infusion to examine tissue loss, neuronal degeneration, and behavioral impairments at 7 days post-stroke, as well as body temperature for 3 days post-stroke. Finally, we evaluated whether bipyridine (25 mg/kg given 2 h prior to surgery and then every 12 h for 3 days) lessens tissue loss, neuronal death, and behavioral deficits after striatal FeCl2 injection. Bipyridine caused a significant hypothermic effect (maximum drop to 34.6 °C for 2-5 h after each injection) in both ICH models; however, in all experiments bipyridine-treated rats were indistinguishable from vehicle controls on all other measures (e.g., tissue loss, behavioral impairments, etc.). These results do not support the use of bipyridine against ICH.
Collapse
|
49
|
Wei S, Sun J, Li J, Wang L, Hall CL, Dix TA, Mohamad O, Wei L, Yu SP. Acute and delayed protective effects of pharmacologically induced hypothermia in an intracerebral hemorrhage stroke model of mice. Neuroscience 2013; 252:489-500. [PMID: 23912033 DOI: 10.1016/j.neuroscience.2013.07.052] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 12/21/2022]
Abstract
Hemorrhagic stroke, including intracerebral hemorrhage (ICH), is a devastating subtype of stroke; yet, effective clinical treatment is very limited. Accumulating evidence has shown that mild to moderate hypothermia is a promising intervention for ischemic stroke and ICH. Current physical cooling methods, however, are less efficient and often impractical for acute ICH patients. The present investigation tested pharmacologically induced hypothermia (PIH) using the second-generation neurotensin receptor (NTR) agonist HPI-201 (formerly known as ABS-201) in an adult mouse model with ICH. Acute or delayed administrations of HPI-201 (2mg/kg bolus injection followed by 2 injections of 1mg/kg, i.p.) were initiated at 1 or 24h after ICH. HPI-201 induced mild hypothermia within 30 min and body and brain temperatures were maintained at 32.7 ± 0.4°C for at least 6h without causing observable shivering. With the 1-h delayed treatment, HPI-201-induced PIH significantly reduced ICH-induced cell death and brain edema compared to saline-treated ICH animals. When HPI-201-induced hypothermia was initiated 24h after the onset of ICH, it still significantly attenuated brain edema, cell death and blood-brain barrier breakdown. HPI-201 significantly decreased the expression of matrix metallopeptidase-9 (MMP-9), reduced caspase-3 activation, and increased Bcl-2 expression in the ICH brain. Moreover, ICH mice received 1-h delayed HPI-201 treatment performed significantly better in the neurological behavior test 48 h after ICH. All together, these data suggest that systemic injection of HPI-201 is an effective hypothermic strategy that protects the brain from ICH injury with a wide therapeutic window. The protective effect of this PIH therapy is partially mediated through the alleviation of apoptosis and neurovascular damage. We suggest that pharmacological hypothermia using the newly developed neurotensin analogs is a promising therapeutic treatment for ICH.
Collapse
Affiliation(s)
- S Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chen-Roetling J, Lu X, Regan KA, Regan RF. A rapid fluorescent method to quantify neuronal loss after experimental intracerebral hemorrhage. J Neurosci Methods 2013; 216:128-36. [PMID: 23583700 PMCID: PMC3679307 DOI: 10.1016/j.jneumeth.2013.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 03/22/2013] [Accepted: 03/24/2013] [Indexed: 10/26/2022]
Abstract
Neuronal loss in tissue surrounding an intracerebral hemorrhage (ICH) is usually quantified by labor-intensive histological methods that are subject to bias. Fluorescent protein expression has been successfully used as a marker of cell viability in vitro and in retinal studies in vivo, but not in any ICH model to date. The potential of this approach was investigated using transgenic mice that constitutively express the red fluorescent protein variant dTomato in central neurons under the control of the Thy1 promoter. Breeding and growth of these mice were similar to their wild-type counterparts; behavioral phenotyping by digital analysis of home cage video recordings detected no differences. Bright fluorescence was evident in fresh brain samples with minimal background fluorescence, and was reduced in tissue surrounding the hematoma. In order to assess fluorescence loss as an injury marker in a planned study, these mice were crossed with heme oxygenase (HO)-2 knockouts and wild-type controls; striatal hemorrhage was induced by stereotactic injection of collagenase. Fluorescence in hemorrhagic striata was reduced to 86.4±3.9%, 62.2±5.1%, and 58.3±3.0% of contra-lateral on days 1, 4 and 8, respectively, and correlated closely with reduction in striatal cell viability as quantified by MTT assay. HO-2 knockout and wild-type values did not differ significantly. Similar results were observed with stereological cell counts of striatal neurons identified by NeuN immunoreactivity. These results suggest that loss of constitutive dTomato fluorescence is an accurate and efficient marker of neuronal loss in tissue surrounding a striatal hematoma.
Collapse
Affiliation(s)
- Jing Chen-Roetling
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, College Building Room 813, Philadelphia, PA 19107
| | - Xiangping Lu
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, College Building Room 813, Philadelphia, PA 19107
| | - Kathleen A. Regan
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, College Building Room 813, Philadelphia, PA 19107
| | - Raymond F. Regan
- Department of Emergency Medicine, Thomas Jefferson University, 1025 Walnut Street, College Building Room 813, Philadelphia, PA 19107
| |
Collapse
|