1
|
Crilly S, Shand I, Bennington A, McMahon E, Flatman D, Tapia VS, Kasher PR. Investigating recovery after a spontaneous intracerebral haemorrhage in zebrafish larvae. Brain Commun 2024; 6:fcae310. [PMID: 39420961 PMCID: PMC11483570 DOI: 10.1093/braincomms/fcae310] [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: 02/28/2024] [Revised: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
Intracerebral haemorrhage is a debilitating stroke sub-type with high morbidity and mortality rates. For survivors, rehabilitation is a long process, and with no available therapeutics to limit the immediate pathophysiology of the haemorrhage, recovery is dependent on individual neuroplasticity. We have previously shown that zebrafish larvae can be used to model spontaneous brain haemorrhage. Zebrafish exhibit innate recovery mechanisms and are often used as a model system for investigation into regeneration after injury, including injury to the nervous system. Here, we investigate the spontaneous and immediate recovery in zebrafish larvae following an intracerebral haemorrhage at 2 days post-fertilisation, during pre-protected stages and over the first 3 weeks of life. We have shown that following the onset of bleed at ∼2 days post-fertilisation zebrafish are capable of clearing the haematoma through the ventricles. Brain cell damage associated with intracerebral haemorrhage is resolved within 48 h, and this recovery is associated with survival rates equal to wildtype and non-haemorrhaged sibling control animals. Larvae express more nestin-positive neural progenitor cells 24 h after injury when the most damage is observed, and through mass spectrometry analysis, we have determined that these cells are highly proliferative and may specially differentiate into oligodendrocytes. This study provides an insight into the haematoma resolution processes in a live, intact organism, and may suggest potential therapeutic approaches to support the recovery of intracerebral haemorrhage patients.
Collapse
Affiliation(s)
- Siobhan Crilly
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester M13 9PT, UK
| | - Isabel Shand
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Abigail Bennington
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester M13 9PT, UK
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Emily McMahon
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester M13 9PT, UK
| | - Daisy Flatman
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester M13 9PT, UK
| | - Victor S Tapia
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester M13 9PT, UK
| | - Paul R Kasher
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
2
|
Zhang Y, Zeng H, Lou F, Tan X, Zhang X, Chen G. SLC45A3 Serves as a Potential Therapeutic Biomarker to Attenuate White Matter Injury After Intracerebral Hemorrhage. Transl Stroke Res 2024; 15:556-571. [PMID: 36913120 PMCID: PMC11106206 DOI: 10.1007/s12975-023-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023]
Abstract
Intracerebral hemorrhage (ICH) is a severe cerebrovascular disease, which impairs patients' white matter even after timely clinical interventions. Indicated by studies in the past decade, ICH-induced white matter injury (WMI) is closely related to neurological deficits; however, its underlying mechanism and pertinent treatment are yet insufficient. We gathered two datasets (GSE24265 and GSE125512), and by taking an intersection among interesting genes identified by weighted gene co-expression networks analysis, we determined target genes after differentially expressing genes in two datasets. Additional single-cell RNA-seq analysis (GSE167593) helped locate the gene in cell types. Furthermore, we established ICH mice models induced by autologous blood or collagenase. Basic medical experiments and diffusion tensor imaging were applied to verify the function of target genes in WMI after ICH. Through intersection and enrichment analysis, gene SLC45A3 was identified as the target one, which plays a key role in the regulation of oligodendrocyte differentiation involving in fatty acid metabolic process, etc. after ICH, and single-cell RNA-seq analysis also shows that it mainly locates in oligodendrocytes. Further experiments verified overexpression of SLC45A3 ameliorated brain injury after ICH. Therefore, SLC45A3 might serve as a candidate therapeutic biomarker for ICH-induced WMI, and overexpression of it may be a potential approach for injury attenuation.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Hanhai Zeng
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Feiyang Lou
- The Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University, Hangzhou, 310020, China
| | - Xiaoxiao Tan
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China
| | - Xiaotong Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- The Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University, Hangzhou, 310020, China.
- College of Electrical Engineering, Zhejiang University, Hangzhou, 310027, China.
- MOE Frontier Science Center for Brain Science and Brain-machine Integration, Zhejiang University, Hangzhou, 310058, China.
| | - Gao Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310016, China.
| |
Collapse
|
3
|
Wu X, Chen Z, Chen Q, Lin C, Zheng X, Yuan B. Nrdp1-mediated Macrophage Phenotypic Regulation Promotes Functional Recovery in Mice with Mild Neurological Impairment after Intracerebral Hemorrhage. Neuroscience 2024; 545:16-30. [PMID: 38431041 DOI: 10.1016/j.neuroscience.2024.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/16/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Neuregulin receptor degradation protein 1 (Nrdp1) is a ring finger E3 ubiquitin ligase involved in some inflammation through ubiquitination, including macrophage polarization following cerebral hemorrhage. However, there is limited understanding regarding the mechanisms through which Nrdp1 modulates macrophage polarization and the potential impact of this modulation on neurological function. Using stereotactic injection and adenoviral transfection techniques, the corresponding animal models were constructed through injecting adenovirus, saline, or blood into the mouse striatum at different periods of time in this research. The alteration in the ratio of various M1/M2 phenotype-associated markers (e.g., CD86, CD206, IL-6, IL-10, etc.) was evaluated through immunohistochemistry, immunofluorescence, western blotting, and elisa assays. Additionally, neurological function scores and behavioral tests were utilized to evaluate changes in neurological function in mice after cerebral hemorrhage. Our results show that overexpression of Nrdp1 promotes the expression of a variety of M2 macrophage-associated markers and enhance transcriptional activity of arginase-1 (Arg1) protein through ubiquitination for early regulation M2 macrophage polarization. Additionally, Nrdp1 promotes hematoma absorption, increases IL-10 expression, inhibits inducible nitric oxide synthase (iNOS), IL-6, and TNF-α production, alleviates neurological impairment and brain edema, and accelerates functional recovery. These findings suggest that modulating macrophage polarization through Nrdp1 could be a therapeutic strategy for neurofunctional impairment in cerebral hemorrhage.
Collapse
Affiliation(s)
- Xiyao Wu
- Department of Neurosurgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian 350000, China
| | - Zhiling Chen
- Department of Neurosurgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian 350000, China
| | - Qiuming Chen
- Department of Neurosurgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian 350000, China
| | - Chuangan Lin
- Department of Neurosurgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian 350000, China
| | - Xiangrong Zheng
- Department of Ophthalmology, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian 350000, China
| | - Bangqing Yuan
- Department of Neurosurgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, Fujian 350000, China; Fuzong Clinical Medical College of Fujian Medical University (900TH Hospital), Fuzhou, Fujian 350000, China.
| |
Collapse
|
4
|
Wang R, Wang J, Zhang Z, Ma B, Sun S, Gao L, Gao G. FGF21 alleviates endothelial mitochondrial damage and prevents BBB from disruption after intracranial hemorrhage through a mechanism involving SIRT6. Mol Med 2023; 29:165. [PMID: 38049769 PMCID: PMC10696847 DOI: 10.1186/s10020-023-00755-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Disruption of the BBB is a harmful event after intracranial hemorrhage (ICH), and this disruption contributes to a series of secondary injuries. We hypothesized that FGF21 may have protective effects after intracranial hemorrhage (ICH) and investigated possible underlying molecular mechanisms. METHODS Blood samples of ICH patients were collected to determine the relationship between the serum level of FGF21 and the [Formula: see text]GCS%. Wild-type mice, SIRT6flox/flox mice, endothelial-specific SIRT6-homozygous-knockout mice (eSIRT6-/- mice) and cultured human brain microvascular endothelial cells (HCMECs) were used to determine the protective effects of FGF21 on the BBB. RESULTS We obtained original clinical evidence from patient data identifying a positive correlation between the serum level of FGF21 and [Formula: see text]GCS%. In mice, we found that FGF21 treatment is capable of alleviating BBB damage, mitigating brain edema, reducing lesion volume and improving neurofunction after ICH. In vitro, after oxyhemoglobin injury, we further explored the protective effects of FGF21 on endothelial cells (ECs), which are a significant component of the BBB. Mitochondria play crucial roles during various types of stress reactions. FGF21 significantly improved mitochondrial biology and function in ECs, as evidenced by alleviated mitochondrial morphology damage, reduced ROS accumulation, and restored ATP production. Moreover, we found that the crucial regulatory mitochondrial factor deacylase sirtuin 6 (SIRT6) played an irreplaceable role in the effects of FGF21. Using endothelial-specific SIRT6-knockout mice, we found that SIRT6 deficiency largely diminished these neuroprotective effects of FGF21. Then, we revealed that FGF21 might promote the expression of SIRT6 via the AMPK-Foxo3a pathway. CONCLUSIONS We provide the first evidence that FGF21 is capable of protecting the BBB after ICH by improving SIRT6-mediated mitochondrial homeostasis.
Collapse
Affiliation(s)
- Runfeng Wang
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Jin Wang
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Zhiguo Zhang
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Bo Ma
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Shukai Sun
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
5
|
Li D, Liu S, Yu T, Liu Z, Sun S, Bragin D, Shirokov A, Navolokin N, Bragina O, Hu Z, Kurths J, Fedosov I, Blokhina I, Dubrovski A, Khorovodov A, Terskov A, Tzoy M, Semyachkina-Glushkovskaya O, Zhu D. Photostimulation of brain lymphatics in male newborn and adult rodents for therapy of intraventricular hemorrhage. Nat Commun 2023; 14:6104. [PMID: 37775549 PMCID: PMC10541888 DOI: 10.1038/s41467-023-41710-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 09/15/2023] [Indexed: 10/01/2023] Open
Abstract
Intraventricular hemorrhage is one of the most fatal forms of brain injury that is a common complication of premature infants. However, the therapy of this type of hemorrhage is limited, and new strategies are needed to reduce hematoma expansion. Here we show that the meningeal lymphatics is a pathway to remove red blood cells from the brain's ventricular system of male human, adult and newborn rodents and is a target for non-invasive transcranial near infrared photobiomodulation. Our results uncover the clinical significance of phototherapy of intraventricular hemorrhage in 4-day old male rat pups that have the brain similar to a preterm human brain. The course of phototherapy in newborn rats provides fast recovery after intraventricular hemorrhage due to photo-improvements of lymphatic drainage and clearing functions. These findings shed light on the mechanisms of phototherapy of intraventricular hemorrhage that can be a clinically relevant technology for treatment of neonatal intracerebral bleedings.
Collapse
Affiliation(s)
- Dongyu Li
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- School of Optical Electronic Information, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
| | - Shaojun Liu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China.
| | - Zhang Liu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
| | - Silin Sun
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
| | - Denis Bragin
- Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA
- Department of Neurology University of New Mexico School of Medicine, Albuquerque, NM, 87131, USA
| | - Alexander Shirokov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Prospekt Entuziastov 13, Saratov, 410049, Russia
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - Nikita Navolokin
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
- Saratov State Medical University, B. Kazachya str., 112, Saratov, 410012, Russia
| | - Olga Bragina
- Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA
| | - Zhengwu Hu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- School of Optical Electronic Information, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
| | - Jürgen Kurths
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
- Physics Department, Humboldt University, Newtonstrasse 15, 12489, Berlin, Germany
- Potsdam Institute for Climate Impact Research, Telegrafenberg A31, 14473, Potsdam, Germany
- Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya 2, building 4, 119435, Moscow, Russia
| | - Ivan Fedosov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - Inna Blokhina
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | | | | | - Andrey Terskov
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - Maria Tzoy
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia
| | - Oxana Semyachkina-Glushkovskaya
- Saratov State University, Astrakhanskaya str., 83, Saratov, 410012, Russia.
- Physics Department, Humboldt University, Newtonstrasse 15, 12489, Berlin, Germany.
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Jia P, Wang J, Ren X, He J, Wang S, Xing Y, Chen D, Zhang X, Zhou S, Liu X, Yu S, Li Z, Jiang C, Zang W, Chen X, Wang J. An enriched environment improves long-term functional outcomes in mice after intracerebral hemorrhage by mechanisms that involve the Nrf2/BDNF/glutaminase pathway. J Cereb Blood Flow Metab 2023; 43:694-711. [PMID: 36635875 PMCID: PMC10108193 DOI: 10.1177/0271678x221135419] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 01/14/2023]
Abstract
Post-stroke depression exacerbates neurologic deficits and quality of life. Depression after ischemic stroke is known to some extent. However, depression after intracerebral hemorrhage (ICH) is relatively unknown. Increasing evidence shows that exposure to an enriched environment (EE) after cerebral ischemia/reperfusion injury has neuroprotective effects in animal models, but its impact after ICH is unknown. In this study, we investigated the effect of EE on long-term functional outcomes in mice subjected to collagenase-induced striatal ICH. Mice were subjected to ICH with the standard environment (SE) or ICH with EE for 6 h/day (8:00 am-2:00 pm). Depressive, anxiety-like behaviors and cognitive tests were evaluated on day 28 with the sucrose preference test, tail suspension test, forced swim test, light-dark transition experiment, morris water maze, and novel object recognition test. Exposure to EE improved neurologic function, attenuated depressive and anxiety-like behaviors, and promoted spatial learning and memory. These changes were associated with increased expression of transcription factor Nrf2 and brain-derived neurotrophic factor (BDNF) and inhibited glutaminase activity in the perihematomal tissue. However, EE did not change the above behavioral outcomes in Nrf2-/- mice on day 28. Furthermore, exposure to EE did not increase BDNF expression compared to exposure to SE in Nrf2-/- mice on day 28 after ICH. These findings indicate that EE improves long-term outcomes in sensorimotor, emotional, and cognitive behavior after ICH and that the underlying mechanism involves the Nrf2/BDNF/glutaminase pathway.
Collapse
Affiliation(s)
- Peijun Jia
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
- School of Life Sciences,
Zhengzhou University, Zhengzhou, China
| | - Junmin Wang
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Xiuhua Ren
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Jinxin He
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Shaoshuai Wang
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Yinpei Xing
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Danyang Chen
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Xinling Zhang
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Siqi Zhou
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Xi Liu
- Department of Neurology,
The First Affiliated Hospital of Zhengzhou University, Zhengzhou,
China
| | - Shangchen Yu
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Zefu Li
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Chao Jiang
- Department of Neurology,
The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou,
China
| | - Weidong Zang
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Xuemei Chen
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| | - Jian Wang
- Department of Anatomy,
School of Basic Medical Sciences, , Zhengzhou
University, Zhengzhou, China
| |
Collapse
|
7
|
Fang J, Song F, Chang C, Yao M. Intracerebral Hemorrhage Models and Behavioral Tests in Rodents. Neuroscience 2023; 513:1-13. [PMID: 36690062 DOI: 10.1016/j.neuroscience.2023.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/08/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023]
Abstract
Intracerebral hemorrhage (ICH) is one of the common types of stroke, which can cause neurological dysfunction. In preclinical ICH studies, researchers often established rodent models by donor/autologous whole blood or a collagenase injection. White matter injury (WMI) can result from primary and secondary injuries after ICH. WMI can lead to short- and long-term neurological impairment, and functional recovery can assess the effect of drug therapy after ICH. Therefore, researchers have devised various behavioral tests to assess dysfunction. This review compares the two ICH modeling methods in rodents and summarizes the pathological mechanisms underlying dysfunction after ICH. We also summarize the functions and characteristics of various behavioral methods, including sensation, motion, emotion, and cognition, to assist researchers in selecting the appropriate tests for preclinical ICH research.
Collapse
Affiliation(s)
- Jie Fang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Fanglai Song
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Chunqi Chang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China.
| | - Min Yao
- School of Pharmaceutical Sciences, Health Science Centre, Shenzhen University, Shenzhen 518060, China; Shenzhen SMQ Group Medical Laboratory, Shenzhen Academy of Measurement and Quality Inspection, Shenzhen 518060, China.
| |
Collapse
|
8
|
Lai JHC, Liu J, Yang T, Huang J, Liu Y, Chen Z, Lee Y, Leung GKK, Chan KWY. Chemical Exchange Saturation Transfer Magnetic Resonance Imaging for Longitudinal Assessment of Intracerebral Hemorrhage and Deferoxamine Treatment at 3T in a Mouse Model. Stroke 2023; 54:255-264. [PMID: 36416125 DOI: 10.1161/strokeaha.122.040830] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Noninvasive imaging of molecular alterations after intracerebral hemorrhage (ICH) could provide valuable information to guide and monitor treatments. Chemical exchange saturation transfer (CEST) magnetic resonance imaging has demonstrated promises in identifying proliferation, necrosis, and changes in cellularity in brain tumors. Here, we applied CEST magnetic resonance imaging to monitor molecular changes in hematoma without and with treatment noninvasively over 2 weeks at 3T using endogenous contrast. METHODS CEST contrast related to proteins at 3.5 ppm (amide proton transfer) and proteins/lipids at -3.5 ppm (relayed nuclear overhauser effect [rNOE]) were examined over 14 days in a collagenase-induced ICH mouse model. Imaging findings were validated with immunohistochemistry based on the ICH neuropathology. We also examined iron-containing phantoms that mimicked iron concentrations in hematoma to ensure the iron will not attenuate the CEST contrast during disease progression. Based on the validity of the CEST contrast of hematoma, we further examined related molecular alterations under iron-chelation treatment with deferoxamine. RESULTS We observed the temporal and spatial differences of CEST contrasts between rNOE at -3.5 ppm and amide proton transfer at 3.5 ppm, in which the core and perihematoma could be identified by rNOE on day 3 and day 14, and amide proton transfer on day 1, day 7, and day 14. Moreover, we observed a 25.7% significant reduction (P<0.05) of rNOE contrast after deferoxamine treatment to the ICH mice on day 3, which was not observable in amide proton transfer contrast. Our histology data indicated that rNOE primarily correlated with the myelin pathology, and amide proton transfer could reflect the cellularity increase at hematoma up to day 7. CONCLUSIONS Significant rNOE changes correlated well with histologic findings, especially myelin lipids, and regional characteristics in hematoma indicate the uniqueness of CEST magnetic resonance imaging in monitoring molecular changes during ICH and treatment.
Collapse
Affiliation(s)
- Joseph H C Lai
- Department of Biomedical Engineering (J.H.C.L., J.H., Y. Liu, Z.C., K.W.Y.C.), City University of Hong Kong
| | - Jiaxin Liu
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong (J.L., T.Y., Y. Liu)
| | - Tian Yang
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong (J.L., T.Y., Y. Liu)
| | - Jianpan Huang
- Department of Biomedical Engineering (J.H.C.L., J.H., Y. Liu, Z.C., K.W.Y.C.), City University of Hong Kong
| | - Yang Liu
- Department of Biomedical Engineering (J.H.C.L., J.H., Y. Liu, Z.C., K.W.Y.C.), City University of Hong Kong.,Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong (J.L., T.Y., Y. Liu)
| | - Zilin Chen
- Department of Biomedical Engineering (J.H.C.L., J.H., Y. Liu, Z.C., K.W.Y.C.), City University of Hong Kong
| | - Youngjin Lee
- Department of Neuroscience (Y. Lee), City University of Hong Kong
| | | | - Kannie W Y Chan
- Department of Biomedical Engineering (J.H.C.L., J.H., Y. Liu, Z.C., K.W.Y.C.), City University of Hong Kong.,Tung Biomedical Sciences Centre (K.W.Y.C.), City University of Hong Kong.,Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD (K.W.Y.C.).,City University of Hong Kong Shenzhen Research Institute, China (K.W.Y.C.).,Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (K.W.Y.C.)
| |
Collapse
|
9
|
Wang J, Wang LJ, Wang LM, Liu ZH, Ren HL, Chen XM, Wang JM, Cai HM, Wei LP, Tian HH. A novel aged mouse model of recurrent intracerebral hemorrhage in the bilateral striatum. Neural Regen Res 2023; 18:344-349. [PMID: 35900428 PMCID: PMC9396476 DOI: 10.4103/1673-5374.346459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The current animal models of stroke primarily model a single intracerebral hemorrhage (ICH) attack, and there is a lack of a reliable model of recurrent ICH. In this study, we established 16-month-old C57BL/6 male mouse models of ICH by injecting collagenase VII-S into the left striatum. Twenty-one days later, we injected collagenase VII-S into the right striatum to simulate recurrent ICH. Our results showed that mice subjected to bilateral striatal hemorrhage had poorer neurological function at the early stage of hemorrhage, delayed recovery in locomotor function, motor coordination, and movement speed, and more obvious emotional and cognitive dysfunction than mice subjected to unilateral striatal hemorrhage. These findings indicate that mouse models of bilateral striatal hemorrhage can well simulate clinically common recurrent ICH. These models should be used as a novel tool for investigating the pathogenesis and treatment targets of recurrent ICH.
Collapse
|
10
|
Vinukonda G, La Gamma EF. Emerging therapies for brain recovery after IVH in neonates: Cord blood derived Mesenchymal Stem Cells (MSC) and Unrestricted Somatic Stem Cells (USSC). Semin Perinatol 2022; 46:151598. [PMID: 35589461 DOI: 10.1016/j.semperi.2022.151598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this report, we summarize evidence on mechanisms of injury after intraventricular hemorrhage resulting in post-hemorrhagic white matter injury and hydrocephalus and correlate that with the possibility of cellular therapy. We describe how two stem cell lines (MSC & USSC) acting in a paracrine fashion offer promise for attenuating the magnitude of injury in animal models and for improved functional recovery by: lowering the magnitude of apoptosis and neuronal cell death, reducing inflammation, and thus, mitigating white matter injury that culminates in improved motor and neurocognitive outcomes. Animal models of IVH are analyzed for their similarity to the human condition and we discuss merits of each approach. Studies on stem cell therapy for IVH in human neonates is described. Lastly, we offer suggestions on what future studies are needed to better understand mechanisms of injury and recovery and argue that human trials need to be expanded in parallel to animal research.
Collapse
Affiliation(s)
- Govindaiah Vinukonda
- Department of Pediatrics, Cell Biology & Anatomy New York Medical College, Valhalla, NY
| | - Edmund F La Gamma
- Department of Pediatrics, Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY.
| |
Collapse
|
11
|
Thangameeran SIM, Pang CY, Lee CH, Tsai ST, Hu WF, Liew HK. Experimental animal models and evaluation techniques in intracerebral hemorrhage. Tzu Chi Med J 2022; 35:1-10. [PMID: 36866349 PMCID: PMC9972928 DOI: 10.4103/tcmj.tcmj_119_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/15/2022] [Accepted: 06/10/2022] [Indexed: 11/04/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is the most lethal type of cerebral stroke without effective therapy. Although clinical trials with various surgeries have been conducted, none have improved clinical outcomes compared to the current medical management for ICH. Several ICH animal models, including autologous blood injection, collagenase injection, thrombin injection, and microballoon inflation methods, have been developed to elucidate the underlying mechanisms of ICH-induced brain injury. These models could also be used for discovering new therapy for ICH preclinically. We summarize the existing ICH animal models and the evaluation parameters used to measure the disease outcomes. We conclude that these models, resembling the different aspects of ICH pathogenesis, have their advantages and disadvantages. None of the current models closely represent the severity of ICH seen in clinical settings. More appropriate models are needed to streamline ICH's clinical outcomes and be used for validating newly developed treatment protocols.
Collapse
Affiliation(s)
| | - Cheng-Yoong Pang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan,Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chien-Hui Lee
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Sheng-Tzung Tsai
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Fen Hu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Hock-Kean Liew
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan,PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan,Address for correspondence: Dr. Hock-Kean Liew, Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, 707, Section 3, Chung-Yang Road, Hualien, Taiwan. E-mail:
| |
Collapse
|
12
|
Metformin Alleviates Delayed Hydrocephalus after Intraventricular Hemorrhage by Inhibiting Inflammation and Fibrosis. Transl Stroke Res 2022; 14:364-382. [PMID: 35852765 DOI: 10.1007/s12975-022-01026-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 01/22/2022] [Accepted: 04/15/2022] [Indexed: 12/28/2022]
Abstract
Intraventricular hemorrhage (IVH) is a subtype of intracerebral hemorrhage (ICH) with high morbidity and mortality. Posthemorrhagic hydrocephalus (PHH) is a common and major complication that affects prognosis, but the mechanism is still unclear. Inflammation and fibrosis have been well established as the major causes of PHH after IVH. In this study, we aimed to investigate the effects of metformin on IVH in adult male mice and further explored the underlying molecular mechanisms of these effects. In the acute phase, metformin treatment exerted dose-dependent neuroprotective effects by reducing periependymal apoptosis and neuronal degeneration and decreasing brain edema. Moreover, high-dose metformin reduced inflammatory cell infiltration and the release of proinflammatory factors, thus protecting ependymal structure integrity and subependymal neurons. In the chronic phase, metformin administration improved neurocognitive function and reduced delayed hydrocephalus. Additionally, metformin significantly inhibited basal subarachnoid fibrosis and ependymal glial scarring. The ependymal structures partially restored. Mechanically, IVH reduced phospho-AMPK (p-AMPK) and SIRT1 expression and activated the phospho-NF-κB (p-NF-κB) inflammatory signaling pathway. However, metformin treatment increased AMPK/SIRT1 expression and lowered the protein expression of p-NF-κB and its downstream inflammation. Compound C and EX527 administration reversed the anti-inflammatory effect of metformin. In conclusion, metformin attenuated neuroinflammation and subsequent fibrosis after IVH by regulating AMPK /SIRT1/ NF-κB pathways, thereby reducing delayed hydrocephalus. Metformin may be a promising therapeutic agent to prevent delayed hydrocephalus following IVH.
Collapse
|
13
|
Li W, Wu LX, Huang BS, Yang LJ, Huang JQ, Li ZS, Jiao J, Cheng T, Li D, Xiong Y. A pilot study: Gut microbiota, metabolism and inflammation in hypertensive intracerebral hemorrhage. J Appl Microbiol 2022; 133:972-986. [PMID: 35560738 DOI: 10.1111/jam.15622] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
AIMS In recent years, the incidence rate of hypertensive intracerebral hemorrhage (HICH) has been increasing, accompanied by high mortality and morbidity, which has brought a heavy burden to the social economy. However, the pathogenesis of HICH is still unclear. This study intends to explore the mechanism of gut microbiota metabolism and inflammation in the process of HICH to provide a theoretical basis for the diagnosis and treatment of HICH. METHODS AND RESULTS HE staining showed that the brain tissues of model group had obvious edema injury, which indicated that the HICH model was successfully constructed. ELISA analysis showed that IL-1β and TNF-α levels in blood and brain tissues were significantly increased, and IL-10 level was significantly decreased in blood. IHC analysis showed that microglia and macrophages were activated in the model group. 16S rRNA sequence showed that the diversity of gut microbiota in HICH patients decreased. And the microbiota belonged to Firmicutes, Proteobacteria and Verrucomicrobia changed significantly. LC-MS/MS analysis showed that the metabolic phenotype of HICH patients changed. And the 3,7-Dimethyluric acid and 7-Methylxanthine related metabolic pathways of caffeine metabolism pathways were down-regulated in patients with HICH. Bacteroides was negatively correlated with the IL-1β and TNF-α level. Blautia was negatively correlated with the IL-1β and TNF-α level, and positively correlated with the IL-10 level. Akkermansia was negatively correlated with the 3,7-Dimethyluric acid and 7-Methylxanthine. CONCLUSION Our study suggested that HICH accompanied by the increased inflammation in peripheral blood and brain, decreased gut microbiota diversity, altered gut metabolic phenotype, and down-regulation of caffeine metabolism pathway. SIGNIFICANCE AND IMPACT OF THE STUDY Our study reported that HICH accompanied by the increased inflammation, decreased gut microbiota diversity, and altered gut metabolic phenotype. Due to the number of patients, this work was a pilot study.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, The First Hospital of Changsha, Changsha, Hunan, China
| | - Li-Xiang Wu
- Department of Physiology, school of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Bai-Sheng Huang
- Department of Physiology, school of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Li-Jian Yang
- Department of Neurosurgery, The First Hospital of Changsha, Changsha, Hunan, China
| | - Jun-Qiang Huang
- Department of Neurosurgery, The First Hospital of Changsha, Changsha, Hunan, China
| | - Zeng-Shi Li
- Department of Neurosurgery, The First Hospital of Changsha, Changsha, Hunan, China
| | - Jia Jiao
- Department of Neurosurgery, The First Hospital of Changsha, Changsha, Hunan, China
| | - Tianxiang Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ding Li
- Department of Interventional vascular surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Yuanyuan Xiong
- Department of Neurosurgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
14
|
Novel targets, treatments, and advanced models for intracerebral haemorrhage. EBioMedicine 2022; 76:103880. [PMID: 35158309 PMCID: PMC8850756 DOI: 10.1016/j.ebiom.2022.103880] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/17/2022] [Accepted: 01/28/2022] [Indexed: 12/20/2022] Open
Abstract
Intracerebral haemorrhage (ICH) is the second most common type of stroke and a major cause of mortality and disability worldwide. Despite advances in surgical interventions and acute ICH management, there is currently no effective therapy to improve functional outcomes in patients. Recently, there has been tremendous progress uncovering new pathophysiological mechanisms underlying ICH that may pave the way for the development of therapeutic interventions. Here, we highlight emerging targets, but also existing gaps in preclinical animal modelling that prevent their exploitation. We particularly focus on (1) ICH aetiology, (2) the haematoma, (3) inflammation, and (4) post-ICH pathology. It is important to recognize that beyond neurons and the brain, other cell types and organs are crucially involved in ICH pathophysiology and successful interventions likely will need to address the entire organism. This review will spur the development of successful therapeutic interventions for ICH and advanced animal models that better reflect its aetiology and pathophysiology.
Collapse
|
15
|
Heidarzadegan AR, Zarifkar A, Sotoudeh N, Namavar MR, Zarifkar AH. Different paradigms of transcranial electrical stimulation improve motor function impairment and striatum tissue injuries in the collagenase-induced intracerebral hemorrhage rat model. BMC Neurosci 2022; 23:6. [PMID: 35093027 PMCID: PMC8801075 DOI: 10.1186/s12868-022-00689-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/19/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
In the horizon of therapeutic restrictions in intracerebral hemorrhage (ICH), recently, non-invasive transcranial electrical stimulation (tES) has achieved considerable prosperities. Translational studies have postulated that transcranial direct current stimulation (tDCS) and the other types of tES remain potentially a novel therapeutic option to reverse or stabilize cognitive and motor impairments.
Objective
The aim of this study was to comparatively evaluate the effects of the four main paradigms of tES, including tDCS, transcranial alternating (tACS), pulsed (tPCS), and random noise (tRNS) stimulations on collagenase-induced sensorimotor impairments and striatum tissue damage in male rats.
Methods
To induce ICH, 0.5 μl of collagenase was injected into the right striatum of male Sprague Dawley rats. One day after surgery, tES, was applied to the animals for seven consecutive days. Motor functions were appraised by neurological deficit score, rotarod, and wire hanging tests on the day before surgery and postoperative days 3, 7, and 14. After behavioral tests, brain tissue was prepared appropriately to perform the stereological evaluations.
Results
The results indicated that the application of the four tES paradigms (tDCS, tACS, tRNS, and tPCS) significantly reversed motor disorders in collagenase-induced ICH groups. Further, the motor function improvement of tACS and tRNS receiving rats in wire-hanging and rotarod tests were higher than the other two tES receiving groups. Structural changes and stereological assessments also confirmed the results of behavioral functions.
Conclusion
Our findings suggest that in addition to tDCS application in the treatment of ICH, other tES paradigms, especially tACS and tRNS may be considered as add-on therapeutic strategies in stroke.
Collapse
|
16
|
Zhu Y, Huang Y, Yang J, Tu R, Zhang X, He WW, Hou CY, Wang XM, Yu JM, Jiang GH. Intranasal insulin ameliorates neurological impairment after intracerebral hemorrhage in mice. Neural Regen Res 2022; 17:210-216. [PMID: 34100458 PMCID: PMC8451559 DOI: 10.4103/1673-5374.314320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In Alzheimer’s disease and ischemic stroke, intranasal insulin can act as a neuroprotective agent. However, whether intranasal insulin has a neuroprotective effect in intracerebral hemorrhage and its potential mechanisms remain poorly understood. In this study, a mouse model of autologous blood-induced intracerebral hemorrhage was treated with 0.5, 1, or 2 IU insulin via intranasal delivery, twice per day, until 24 or 72 hours after surgery. Compared with saline treatment, 1 IU intranasal insulin treatment significantly reduced hematoma volume and brain edema after cerebral hemorrhage, decreased blood-brain barrier permeability and neuronal degeneration damage, reduced neurobehavioral deficits, and improved the survival rate of mice. Expression levels of p-AKT and p-GSK3β were significantly increased in the perihematoma tissues after intranasal insulin therapy. Our findings suggest that intranasal insulin therapy can protect the neurological function of mice after intracerebral hemorrhage through the AKT/GSK3β signaling pathway. The study was approved by the Ethics Committee of the North Sichuan Medical College of China (approval No. NSMC(A)2019(01)) on January 7, 2019.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Yi Huang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Jin Yang
- Department of Intensive Care Unit, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Rong Tu
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Xin Zhang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Wei-Wei He
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Chang-Yue Hou
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Xiao-Ming Wang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Ju-Ming Yu
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| | - Guo-Hui Jiang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College; Institute of Neurological Diseases, North Sichuan Medical College, Nanchong, Sichuan Province, China
| |
Collapse
|
17
|
Zhu W, Gao Y, Chang CF, Wan JR, Zhu SS, Wang J. Correction: Mouse Models of Intracerebral Hemorrhage in Ventricle, Cortex, and Hippocampus by Injections of Autologous Blood or Collagenase. PLoS One 2021; 16:e0261640. [PMID: 34910780 PMCID: PMC8673673 DOI: 10.1371/journal.pone.0261640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
18
|
O-GlcNAcase inhibitor has protective effects in intracerebral hemorrhage by suppressing the inflammatory response. Neuroreport 2021; 32:1349-1356. [PMID: 34718246 DOI: 10.1097/wnr.0000000000001734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is aggravated by immune cells that participate in the inflammatory response from the blood-brain barrier (BBB). O-Glycosylation has been reported to regulate the inflammatory response in the central nervous system but its cerebral protective effects remain unknown. Therefore, this study was carried out to investigate the protective effects of O-GlcNAcylation in a murine model of ICH and the possible mechanisms involved. METHODS The effects of O-GlcNAcylation on hematoma and edema formation were tested using pathological and dry/wet weight methods, whereas its effects on neural function were determined using neurologic tests. The effect of O-GlcNAcylation on BBB integrity was determined by Evans blue dye extrusion. Flow cytometry was used to quantify the immune cells in the central nervous system. Immunofluorescence was used to detect the protective effect of O-GlcNAcylation in ICH. RESULTS The hematoma volume was significantly lower in the prevention and treatment groups than in the control group after ICH induction, indicating that O-GlcNAcylation had reduced the formation of cerebral hematoma in ICH. In the prevention and treatment groups, the modified neurological severity score, corner turn test and rotating rod test results were improved and the BBB integrity was better than that in the control group. O-GlcNAcylation also regulated the microglia, neutrophils and other central nervous system immune cells after ICH, effectively reducing the inflammatory response. CONCLUSIONS O-GlcNAcylation played an important role in suppressing the inflammatory response, enhancing the BBB integrity and reducing edema after ICH.
Collapse
|
19
|
Mehmood Siddiqui E, Mehan S, Upadhayay S, Khan A, Halawi M, Ahmed Halawi A, Alsaffar RM. Neuroprotective efficacy of 4-Hydroxyisoleucine in experimentally induced intracerebral hemorrhage. Saudi J Biol Sci 2021; 28:6417-6431. [PMID: 34764759 PMCID: PMC8568986 DOI: 10.1016/j.sjbs.2021.07.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a severe form of brain injury, which is a major cause of mortality in humans. Hydrocephalus and cerebral hematoma lead to severe neurological deficits. A single autologous blood (ALB) injection in rats' brains induces hemorrhage and other conditions that regularly interfere with the standard treatment of several cellular and molecular pathways. Several studies have found that IGF-1/GLP-1 decreases the production of inflammatory markers in peripheral tissues, while some have found that they also have pro-inflammatory functions. Since these receptors are down-regulated in hemorrhagic situations, we looked into the potential neuroprotective effect of 4-hydroxyisoleucine (4-HI); 50 mg/kg and 100 mg/kg, an active compound Trigonellafoenum-graecum, on post-hemorrhagic deficits in rats. Long-term oral administration of 4-HI for 35 days has improved behavioral and neurochemical deficits and severe pathological changes and improved cellular and molecular markers, apoptotic markers in the ALB-induced ICH experimental model. Furthermore, the findings revealed that 4-HI also improved the levels of other neurotransmitters (Ach, DOPA, GABA, glutamate); inflammatory cytokines (TNF-alpha, IL-1β, IL-17), and oxidative stress markers (MDA, nitrite, LDH, AchE, SOD, CAT, GPx, GSH) in the brain when evaluated after Day 35. There is no proven treatment available for the prevention of post-brain hemorrhage and neurochemical malfunction; available therapy is only for symptomatic relief of the patient. Thus, 4-HI could be a potential clinical approach for treating post-brain haemorrhage and neurochemical changes caused by neurological damage. Furthermore, 4-HI may be linked to other standard therapeutic therapies utilized in ICH as a potential pharmacological intervention.
Collapse
Affiliation(s)
- Ehraz Mehmood Siddiqui
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shubham Upadhayay
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Maryam Halawi
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | - Rana M Alsaffar
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, P.O.Box-173, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
20
|
Chen W, Jiang L, Hu Y, Fang G, Yang B, Li J, Liang N, Wu L, Hussain Z. Nanomedicines, an emerging therapeutic regimen for treatment of ischemic cerebral stroke: A review. J Control Release 2021; 340:342-360. [PMID: 34695522 DOI: 10.1016/j.jconrel.2021.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022]
Abstract
Owing to its intricate pathophysiology, cerebral stroke is a serious medical condition caused by interruption or obstruction of blood supply (blockage of vasculature) to the brain tissues which results in diminished supply of essential nutrients and oxygen (hypoxia) and ultimate necrosis of neuronal tissues. A prompt risks assessment and immediate rational therapeutic plan with proficient neuroprotection play critically important role in the effective management of this neuronal emergency. Various conventional medications are being used for treatment of acute ischemic cerebral stroke but fibrinolytic agents, alone or in combination with other agents are considered the mainstay. These clot-busting agents effectively restore blood supply (reperfusion) to ischemic regions of the brain; however, their clinical significance is hampered due to various factors such as short plasma half-life, limited distribution to brain tissues due to the presence of highly efficient physiological barrier, blood brain barrier (BBB), and lacking of target-specific delivery to the ischemic brain regions. To alleviate these issues, various types of nanomedicines such as polymeric nanoparticles (NPs), liposomes, nanoemulsion, micelles and dendrimers have been designed and evaluated. The implication of these newer therapies (nanomedicines) have revolutionized the therapeutic outcomes by improving the plasma half-life, permeation across BBB, efficient distribution to ischemic cerebral tissues and neuroprotection. Furthermore, the adaptation of some diverse techniques including PEGylation, tethering of targeting ligands on the surfaces of nanomedicines, and pH responsive features have also been pondered. The implication of these emerging adaptations have shown remarkable potential in maximizing the targeting efficiency of drugs to ischemic brain tissues, simultaneous delivery of drugs and imaging agents (for early prognosis as well as monitoring of therapy), and therapeutic outcomes such as long-term neuroprotection.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Lingfei Jiang
- Graduate College, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Yueqiang Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China.
| | - Gang Fang
- Guangxi Zhuang and Yao Medicine Engineering Technology Research Center, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Bilin Yang
- Graduate College, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Junhong Li
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Ni Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Lin Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China.
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
21
|
Zhang R, Wang J, Huang L, Wang TJ, Huang Y, Li Z, He J, Sun C, Wang J, Chen X, Wang J. The pros and cons of motor, memory, and emotion-related behavioral tests in the mouse traumatic brain injury model. Neurol Res 2021; 44:65-89. [PMID: 34308784 DOI: 10.1080/01616412.2021.1956290] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a medical emergency with high morbidity and mortality. Motor, memory, and emotion-related deficits are common symptoms following TBI, yet treatment is very limited. To develop new drugs and find new therapeutic avenues, a wide variety of TBI models have been established to mimic the heterogeneity of TBI. In this regard, along with histologic measures, behavioral functional outcomes provide valuable insight into the underlying neuropathology and guide neurorehabilitation efforts for neuropsychiatric impairment after TBI. Development, characterization, and application of behavioral tests that can assess functional neurologic deficits are essential to the development of translational therapies. This comprehensive review aims to summarize 19 common behavioral tests from three aspects (motor, memory, and emotion-related) that are associated with TBI pathology. Discussion covers the apparatus, the test steps, the evaluation indexes, data collection and analysis, animal performance and applications, advantages and disadvantages as well as precautions to eliminate bias wherever possible. We discussed recent studies on TBI-related preconditioning, biomarkers, and optimized behavioral protocols. The neuropsychologic tests employed in clinics were correlated with those used in mouse TBI models. In summary, this review provides a comprehensive, up-to-date reference for TBI researchers to choose the right neurobehavioral protocol according to the research objectives of their translational investigation.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junming Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Yinrou Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zefu Li
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinxin He
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chen Sun
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Shi X, Bai H, Wang J, Wang J, Huang L, He M, Zheng X, Duan Z, Chen D, Zhang J, Chen X, Wang J. Behavioral Assessment of Sensory, Motor, Emotion, and Cognition in Rodent Models of Intracerebral Hemorrhage. Front Neurol 2021; 12:667511. [PMID: 34220676 PMCID: PMC8248664 DOI: 10.3389/fneur.2021.667511] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/20/2021] [Indexed: 11/15/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is the second most common type of stroke and has one of the highest fatality rates of any disease. There are many clinical signs and symptoms after ICH due to brain cell injury and network disruption resulted from the rupture of a tiny artery and activation of inflammatory cells, such as motor dysfunction, sensory impairment, cognitive impairment, and emotional disturbance, etc. Thus, researchers have established many tests to evaluate behavioral changes in rodent ICH models, in order to achieve a better understanding and thus improvements in the prognosis for the clinical treatment of stroke. This review summarizes existing protocols that have been applied to assess neurologic function outcomes in the rodent ICH models such as pain, motor, cognition, and emotion tests. Pain tests include mechanical, hot, and cold pain tests; motor tests include the following 12 types: neurologic deficit scale test, staircase test, rotarod test, cylinder test, grid walk test, forelimb placing test, wire hanging test, modified neurologic severity score, beam walking test, horizontal ladder test, and adhesive removal test; learning and memory tests include Morris water maze, Y-maze, and novel object recognition test; emotion tests include elevated plus maze, sucrose preference test, tail suspension test, open field test, and forced swim test. This review discusses these assessments by examining their rationale, setup, duration, baseline, procedures as well as comparing their pros and cons, thus guiding researchers to select the most appropriate behavioral tests for preclinical ICH research.
Collapse
Affiliation(s)
- Xiaoyu Shi
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huiying Bai
- Zhengzhou University Hospital Outpatient Surgery Center, Zhengzhou, China
| | - Junmin Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiarui Wang
- Keieger School of Arts and Sciences, The Johns Hopkins University, Baltimore, MD, United States
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Meimei He
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xuejun Zheng
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zitian Duan
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Danyang Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiaxin Zhang
- Saint John Paul the Great Catholic High School, Dumfries, VA, United States
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jian Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
23
|
Minj E, Upadhayay S, Mehan S. Nrf2/HO-1 Signaling Activator Acetyl-11-keto-beta Boswellic Acid (AKBA)-Mediated Neuroprotection in Methyl Mercury-Induced Experimental Model of ALS. Neurochem Res 2021; 46:2867-2884. [PMID: 34075522 DOI: 10.1007/s11064-021-03366-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/28/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Methylmercury (MeHg) is a potent neurotoxin that causes neurotoxicity and neuronal cell death. MeHg exposure also leads to oligodendrocyte destruction, glial cell overactivation, and demyelination of motor neurons in the motor cortex and spinal cord. As a result, MeHg plays an important role in the progression of amyotrophic lateral sclerosis (ALS)-like neurocomplications. ALS is a fatal neurodegenerative disorder in which neuroinflammation is the leading cause of further CNS demyelination. Nuclear factor erythroid-2-related factor-2 (Nrf2)/Heme oxygenase-1 (HO-1) signaling pathway was thought to be a potential target for neuroprotection in ALS. Acetyl-11-keto-beta-boswellic acid (AKBA) is a multi-component pentacyclic triterpenoid mixture derived from Boswellia serrata with anti-inflammatory and antioxidant properties. The research aimed to investigate whether AKBA, as a Nrf2 / HO-1 activator, can provide protection against ALS. Thus, we explored the role of AKBA on the Nrf2/HO-1 signaling pathway in a MeHg-induced experimental ALS model. In this study, ALS was induced in Wistar rats by oral gavage of MeHg 5 mg/kg for 21 days. An open field test, force swim test, and grip strength were performed to observe experimental rats' motor coordination behaviors. In contrast, a morris water maze was performed for learning and memory. Administration of AKBA 50 mg/kg and AKBA 100 mg/kg continued from day 22 to 42. Neurochemical parameters were evaluated in the rat's brain homogenate. In the meantime, post-treatment with AKBA significantly improved behavioral, neurochemical, and gross pathological characteristics in the brain of rats by increasing the amount of Nrf2/HO-1 in brain tissue. Collectively, our findings indicated that AKBA could potentially avoid demyelination and encourage remyelination.
Collapse
Affiliation(s)
- Elizabeth Minj
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Shubham Upadhayay
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
24
|
Purohit D, Finkel DA, Malfa A, Liao Y, Ivanova L, Kleinman GM, Hu F, Shah S, Thompson C, Joseph E, Wolin MS, Cairo MS, La Gamma EF, Vinukonda G. Human Cord Blood Derived Unrestricted Somatic Stem Cells Restore Aquaporin Channel Expression, Reduce Inflammation and Inhibit the Development of Hydrocephalus After Experimentally Induced Perinatal Intraventricular Hemorrhage. Front Cell Neurosci 2021; 15:633185. [PMID: 33897371 PMCID: PMC8062878 DOI: 10.3389/fncel.2021.633185] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Intraventricular hemorrhage (IVH) is a severe complication of preterm birth associated with cerebral palsy, intellectual disability, and commonly, accumulation of cerebrospinal fluid (CSF). Histologically, IVH leads to subependymal gliosis, fibrosis, and disruption of the ependymal wall. Importantly, expression of aquaporin channels 1 and 4 (AQP1 and AQP4) regulating respectively, secretion and absorption of cerebrospinal fluids is altered with IVH and are associated with development of post hemorrhagic hydrocephalus. Human cord blood derived unrestricted somatic stem cells (USSCs), which we previously demonstrated to reduce the magnitude of hydrocephalus, as having anti-inflammatory, and beneficial behavioral effects, were injected into the cerebral ventricles of rabbit pups 18 h after glycerol-induced IVH. USSC treated IVH pups showed a reduction in ventricular size when compared to control pups at 7 and 14 days (both, P < 0.05). Histologically, USSC treatment reduced cellular infiltration and ependymal wall disruption. In the region of the choroid plexus, immuno-reactivity for AQP1 and ependymal wall AQP4 expression were suppressed after IVH but were restored following USSC administration. Effects were confirmed by analysis of mRNA from dissected choroid plexus and ependymal tissue. Transforming growth factor beta (TGF-β) isoforms, connective tissue growth factor (CTGF) and matrix metalloprotease-9 (MMP-9) mRNA, as well as protein levels, were significantly increased following IVH and restored towards normal with USSC treatment (P < 0.05). The anti-inflammatory cytokine Interleukin-10 (IL-10) mRNA was reduced in IVH, but significantly recovered after USSC injection (P < 0.05). In conclusion, USSCs exerted anti-inflammatory effects by suppressing both TGF-β specific isoforms, CTGF and MMP-9, recovered IL-10, restored aquaporins expression towards baseline, and reduced hydrocephalus. These results support the possibility of the use of USSCs to reduce IVH consequences in prematurity.
Collapse
Affiliation(s)
- Deepti Purohit
- The Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Dina A Finkel
- The Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Ana Malfa
- The Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Larisa Ivanova
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - George M Kleinman
- Department of Pathology, Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Furong Hu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Shetal Shah
- The Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, United States
| | - Carl Thompson
- Department of Physiology, New York Medical College, Valhalla, NY, United States
| | - Etlinger Joseph
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, NY, United States
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States.,Departments of Medicine, Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Edmund F La Gamma
- The Regional Neonatal Center, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, Valhalla, NY, United States.,Department of Pediatrics, New York Medical College, Valhalla, NY, United States.,Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | - Govindaiah Vinukonda
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States.,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
25
|
Bahadar GA, Shah ZA. Intracerebral Hemorrhage and Diabetes Mellitus: Blood-Brain Barrier Disruption, Pathophysiology, and Cognitive Impairments. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:312-326. [PMID: 33622232 DOI: 10.2174/1871527320666210223145112] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/15/2020] [Accepted: 11/02/2020] [Indexed: 11/22/2022]
Abstract
There is a surge in diabetes incidence with an estimated 463 million individuals been diagnosed worldwide. Diabetes Mellitus (DM) is a major stroke-related comorbid condition that increases the susceptibility of disabling post-stroke outcomes. Although less common, intracerebral hemorrhage (ICH) is the most dramatic subtype of stroke that is associated with higher mortality, particularly in DM population. Previous studies have focused mainly on the impact of DM on ischemic stroke. Few studies have focused on impact of DM on ICH and discussed the blood-brain barrier disruption, brain edema, and hematoma formation. However, more recently, investigating the role of oxidative damage and reactive oxygen species (ROS) production in preclinical studies involving DM-ICH animal models has gained attention. But, little is known about the correlation between neuroinflammatory processes, glial cells activation, and peripheral immune cell invasion with DM-ICH injury. DM and ICH patients experience impaired abilities in multiple cognitive domains by relatively comparable mechanisms, which could get exacerbated in the setting of comorbidities. In this review, we discuss both the pathology of DM as a comorbid condition for ICH and the potential molecular therapeutic targets for the clinical management of the ICH and its recovery.
Collapse
Affiliation(s)
- Ghaith A Bahadar
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614. United States
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614. United States
| |
Collapse
|
26
|
Abstract
Rodents are the most widely used experimental animals in stroke research due to their similar vascular anatomy, high reproductive rates, and availability of transgenic models. However, the difficulties in assessing higher brain functions, such as cognition and memory, in rodents decrease the translational potential of these studies. In this review, we summarize commonly used motor/sensorimotor and cognition tests in rodent models of stroke. Specifically, we first briefly introduce the objective and procedure of each behavioral test. Next, we summarize the application of each test in both ischemic stroke and hemorrhagic stroke. Last, the advantages and disadvantages of these tests in assessing stroke outcome are discussed. This review summarizes commonly used behavioral tests in stroke studies and compares their applications in different stroke types.
Collapse
Affiliation(s)
- Jingsong Ruan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA, USA
| |
Collapse
|
27
|
Romantsik O, Bruschettini M, Ley D. Intraventricular Hemorrhage and White Matter Injury in Preclinical and Clinical Studies. Neoreviews 2020; 20:e636-e652. [PMID: 31676738 DOI: 10.1542/neo.20-11-e636] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Germinal matrix-intraventricular hemorrhage (IVH) occurs in nearly half of infants born at less than 26 weeks' gestation. Up to 50% of survivors with IVH develop cerebral palsy, cognitive deficits, behavioral disorders, posthemorrhagic ventricular dilatation, or a combination of these sequelae. After the initial bleeding and the primary brain injury, inflammation and secondary brain injury might lead to periventricular leukomalacia or diffuse white matter injury. Potential factors that are involved include microglia and astrocyte activation, degradation of blood components with release of "toxic" products, infiltration of the brain by systemic immune cells, death of neuronal and glial cells, and arrest of preoligodendrocyte maturation. In addition, impairment of the blood-brain barrier may play a major role in the pathophysiology. A wide range of animal models has been used to explore causes and mechanisms leading to IVH-induced brain injury. Preclinical studies have identified potential targets for enhancing brain repair. However, little has been elucidated about the effectiveness of potential interventions in clinical studies. A systematic review of available preclinical and clinical studies might help identify research gaps and which types of interventions may be prioritized. Future trials should report clinically robust and long-term outcomes after IVH.
Collapse
Affiliation(s)
- Olga Romantsik
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
28
|
Yan J, Zuo G, Sherchan P, Huang L, Ocak U, Xu W, Travis ZD, Wang W, Zhang JH, Tang J. CCR1 Activation Promotes Neuroinflammation Through CCR1/TPR1/ERK1/2 Signaling Pathway After Intracerebral Hemorrhage in Mice. Neurotherapeutics 2020; 17:1170-1183. [PMID: 31898284 PMCID: PMC7609528 DOI: 10.1007/s13311-019-00821-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The activation of C-C chemokine receptor type 1 (CCR1) has been shown to be pro-inflammatory in several animal models of neurological diseases. The objective of this study was to investigate the activation of CCR1 on neuroinflammation in a mouse model of intracerebral hemorrhage (ICH) and the mechanism of CCR1/tetratricopeptide repeat 1 (TPR1)/extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway in CCR1-mediated neuroinflammation. Adult male CD1 mice (n = 210) were used in the study. The selective CCR1 antagonist Met-RANTES was administered intranasally at 1 h after autologous blood injection. To elucidate potential mechanism, a specific ERK1/2 activator (ceramide C6) was administered prior to Met-RANTES treatment; CCR1 activator (recombinant CCL5, rCCL5) and TPR1 CRISPR were administered in naïve mouse. Neurobehavioral assessments, brain water content, immunofluorescence staining, and western blot were performed. The endogenous expressions of CCR1, CCL5, TPR1, and p-ERK1/2 were increased in the brain after ICH. CCR1 were expressed on microglia, neurons, and astrocytes. The inhibition of CCR1 with Met-RANTES improved neurologic function, decreased brain edema, and suppressed microglia/macrophage activations and neutrophil infiltration after ICH. Met-RANTES treatment decreased expressions of CCR1, TPR1, p-ERK, TNF-α, and IL-1β, which was reversed by ceramide C6. The brain CCR1 activation by rCCL5 injection in naïve mouse resulted in neurological deficits and increased expressions of CCR1, TPR1, p-ERK, TNF-α, and IL-1β. These detrimental effects of rCCL5 were reversed by TPR1 knockdown using TPR1 CRISPR. Our study demonstrated that CCR1 activation promoted neuroinflammation through CCR1/TPR1/ERK1/2 signaling pathway after ICH in mice. CCR1 inhibition with Met-RANTES attenuated neuroinflammation, thereby reducing brain edema and improving neurobehavioral functions. Targeting CCR1 activation may provide a promising therapeutic approach in the management of ICH patients.
Collapse
Affiliation(s)
- Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
- Department of Neurosurgery, The Affiliated Taicang Hospital, Soochow University, Taicang, Suzhou, 215400, Jiangsu, China
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Umut Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Weilin Xu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - Zachary D Travis
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
- Department of Earth and Biological Sciences, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Wenna Wang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, 11041 Campus Street, Loma Linda, CA, 92354, USA.
| |
Collapse
|
29
|
miR-183-5p alleviates early injury after intracerebral hemorrhage by inhibiting heme oxygenase-1 expression. Aging (Albany NY) 2020; 12:12869-12895. [PMID: 32602850 PMCID: PMC7377845 DOI: 10.18632/aging.103343] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
Differences in microRNA (miRNA) expression after intracerebral hemorrhage (ICH) have been reported in human and animal models, and miRNAs are being investigated as a new treatment for inflammation and oxidative stress after ICH. In this study, we found that microRNA-183-5p expression was decreased in the mouse brain after ICH. To investigate the effect of miRNA-183-5p on injury and repair of brain tissue after ICH, saline, miRNA-183-5p agomir, or miRNA-183-5p antagomir were injected into the lateral ventricles of 8-week-old mice with collagenase-induced ICH. Three days after ICH, mice treated with exogenous miRNA-183-5p showed less brain edema, neurobehavioral defects, inflammation, oxidative stress, and ferrous deposition than control mice. In addition, by alternately treating mice with a heme oxygenase-1 (HO-1) inducer, a HO-1 inhibitor, a nuclear factor erythroid 2-related factor (Nrf2) activator, and Nrf2 knockout, we demonstrated an indirect, HO-1-dependent regulatory relationship between miRNA-183-5p and Nrf2. Our results indicate that miRNA-183-5p and HO-1 are promising therapeutic targets for controlling inflammation and oxidative damage after hemorrhagic stroke.
Collapse
|
30
|
miR-331-3p Inhibits Inflammatory Response after Intracerebral Hemorrhage by Directly Targeting NLRP6. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6182464. [PMID: 32596340 PMCID: PMC7298275 DOI: 10.1155/2020/6182464] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/18/2020] [Indexed: 01/02/2023]
Abstract
Background The mechanism of inflammatory reaction after intracerebral hemorrhage remains unclear, which to some extent restrains the therapeutic development of hemorrhagic stroke. The present study attempts to verify whether NLRP6 plays an important role in inflammatory reaction after intracerebral hemorrhage and identify the critical microRNA during the process. Methods Suitable simulated cerebral hemorrhage environments were established in vitro and in vivo. BV2 cells were treated with hemin to induce cell damage. Collagenase was used to establish a model of mouse cerebral hemorrhage. The relationship among NLRP6, miR-331-3p, and the corresponding inflammatory expression was closely observed during this process. Techniques, such as western blot, real-time quantitative PCR, immunofluorescence, and immunocytochemistry, were used to detect the expression of relative genes and molecules in the in vitro and in vivo models. Results Downregulated miR-331-3p increased the expression of NLRP6 and alleviated the expression of TNF-α and IL-6. The neurological function recovery of mice was promoted after intracerebral hemorrhage. Conclusion miR-331-3p regulated the inflammatory response after cerebral hemorrhage by negatively regulating the expression of NLRP6.
Collapse
|
31
|
Biological Effects of Hydrogen Sulfide and Its Protective Role in Intracerebral Hemorrhage. J Mol Neurosci 2020; 70:2020-2030. [DOI: 10.1007/s12031-020-01608-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022]
|
32
|
Rajdev K, Siddiqui EM, Jadaun KS, Mehan S. Neuroprotective potential of solanesol in a combined model of intracerebral and intraventricular hemorrhage in rats. IBRO Rep 2020; 8:101-114. [PMID: 32368686 PMCID: PMC7184235 DOI: 10.1016/j.ibror.2020.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Intracerebral hemorrhage (ICH) may be caused by trauma, aneurysm and arteriovenous malformation, as can any bleeding within the intracranial vault, including brain parenchyma and adjacent meningeal spaces (aneurism and atreovenous malformation). ICH is the cerebral stroke with the least treatable form. Over time, intraventricular hemorrhage (IVH) is associated with ICH, which contributes to hydrocephalus, and the major cause of most hemorrhagic death (Due to the cerebral hemorrhage and post hemorrhagic surgeries). Most patients suffer from memory impairment, grip strength, posture, and cognitive dysfunctions attributable to cerebral hemorrhage or post-brain hemorrhagic surgery. Nevertheless, a combined model of ICH based IVH is not present pre-clinically. Autologous blood (ALB) injection (20 μl/5 min) in the rat brain triggers hemorrhage, such as factors that further interfere with the normal functioning of neuroinflammatory cytokines, oxidative stress, and neurotransmitter dysfunction, such as CoQ10 insufficiency and dysregulation of mitochondrial ETC-complexes. For the prevention of post-brain hemorrhagic behavioral and neurochemical dysfunctions, there is no specific drug treatment available, only available therapy used to provide symptomatic relief. The current study reveals that long-term administration of Solanesol (SNL) 40 and 60 mg/kg alone and in combination with available drug therapy Donepezil (DNP) 3 mg/kg, Memantine (MEM) 20 mg/kg, Celecoxib (CLB) 20 mg/kg, Pregabalin (PGB) 30 mg/kg, may provide the neuroprotective effect by improving behavioral and neurochemical deficits, and gross pathological changes in ALB induced combined experimental model of ICH-IVH in post brain hemorrhagic conditions in rats. Thus, SNL can be a potential therapeutic approach to improve neuronal mitochondrial dysfunction associated with post brain hemorrhagic behavioral and neurochemical alterations.
Collapse
Affiliation(s)
- Kajal Rajdev
- Neuropharmacology Division, ISF College of Pharmacy, Moga, 142001 Punjab, India
| | | | | | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, 142001 Punjab, India
| |
Collapse
|
33
|
Karimy JK, Reeves BC, Damisah E, Duy PQ, Antwi P, David W, Wang K, Schiff SJ, Limbrick DD, Alper SL, Warf BC, Nedergaard M, Simard JM, Kahle KT. Inflammation in acquired hydrocephalus: pathogenic mechanisms and therapeutic targets. Nat Rev Neurol 2020; 16:285-296. [PMID: 32152460 DOI: 10.1038/s41582-020-0321-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
Hydrocephalus is the most common neurosurgical disorder worldwide and is characterized by enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles resulting from failed CSF homeostasis. Since the 1840s, physicians have observed inflammation in the brain and the CSF spaces in both posthaemorrhagic hydrocephalus (PHH) and postinfectious hydrocephalus (PIH). Reparative inflammation is an important protective response that eliminates foreign organisms, damaged cells and physical irritants; however, inappropriately triggered or sustained inflammation can respectively initiate or propagate disease. Recent data have begun to uncover the molecular mechanisms by which inflammation - driven by Toll-like receptor 4-regulated cytokines, immune cells and signalling pathways - contributes to the pathogenesis of hydrocephalus. We propose that therapeutic approaches that target inflammatory mediators in both PHH and PIH could address the multiple drivers of disease, including choroid plexus CSF hypersecretion, ependymal denudation, and damage and scarring of intraventricular and parenchymal (glia-lymphatic) CSF pathways. Here, we review the evidence for a prominent role of inflammation in the pathogenic mechanism of PHH and PIH and highlight promising targets for therapeutic intervention. Focusing research efforts on inflammation could shift our view of hydrocephalus from that of a lifelong neurosurgical disorder to that of a preventable neuroinflammatory condition.
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Eyiyemisi Damisah
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Phan Q Duy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Prince Antwi
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Wyatt David
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Kevin Wang
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Steven J Schiff
- Departments of Neurosurgery, Engineering Science & Mechanics, and Physics; Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| | - David D Limbrick
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Benjamin C Warf
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.,Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology and Yale-Rockefeller NIH Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
34
|
Reeves BC, Karimy JK, Kundishora AJ, Mestre H, Cerci HM, Matouk C, Alper SL, Lundgaard I, Nedergaard M, Kahle KT. Glymphatic System Impairment in Alzheimer's Disease and Idiopathic Normal Pressure Hydrocephalus. Trends Mol Med 2020; 26:285-295. [PMID: 31959516 PMCID: PMC7489754 DOI: 10.1016/j.molmed.2019.11.008] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/11/2019] [Accepted: 11/20/2019] [Indexed: 12/31/2022]
Abstract
Approximately 10% of dementia patients have idiopathic normal pressure hydrocephalus (iNPH), an expansion of the cerebrospinal fluid (CSF)-filled brain ventricles. iNPH and Alzheimer's disease (AD) both exhibit sleep disturbances, build-up of brain metabolic wastes and amyloid-β (Aβ) plaques, perivascular reactive astrogliosis, and mislocalization of astrocyte aquaporin-4 (AQP4). The glia-lymphatic (glymphatic) system facilitates brain fluid clearance and waste removal during sleep via glia-supported perivascular channels. Human studies have implicated impaired glymphatic function in both AD and iNPH. Continued investigation into the role of glymphatic system biology in AD and iNPH models could lead to new strategies to improve brain health by restoring homeostatic brain metabolism and CSF dynamics.
Collapse
Affiliation(s)
- Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Adam J Kundishora
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Humberto Mestre
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - H Mert Cerci
- Istanbul Universty-Cerrahpasa, Cerrahpasa School of Medicine, Istanbul 34096, Turkey
| | - Charles Matouk
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06510, USA
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Iben Lundgaard
- Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular and Molecular Physiology; and Yale-Rockefeller National Institutes of Health (NIH) Centers for Mendelian Genomics, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
35
|
Li W, Li L, Li W, Chopp M, Venkat P, Zacharek A, Chen Z, Landschoot-Ward J, Chen J. Spleen associated immune-response mediates brain-heart interaction after intracerebral hemorrhage. Exp Neurol 2020; 327:113209. [PMID: 31987832 DOI: 10.1016/j.expneurol.2020.113209] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/06/2020] [Accepted: 01/24/2020] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND PURPOSE Intracerebral hemorrhage (ICH) patients frequently encounter cardiovascular complications which may contribute to increased mortality and poor long term outcome. ICH induces systemic oxidative stress and activates peripheral immune responses which are involved in the pathological cascade leading to cardiac dysfunction and heart failure after ICH. We have previously reported that ICH induces progressive cardiac dysfunction in mice without primary cardiac diseases. In this study, we have investigated the role of immune response in mediating cardiac dysfunction post ICH in mice. METHODS Adult male C57BL/6 J mice were randomly assigned to the following groups (n = 8/group): 1) sham control; 2) ICH; 3) splenectomy with ICH (ICH + Spx); 4) splenectomy alone (Spx). Echocardiography was performed at 7 and 28 days after ICH. A battery of neurological and cognitive tests were performed. Flow cytometry, western blot and immunostaining were used to test mechanisms of ICH induced cardiac dysfunction. RESULTS Compared to sham control mice, Spx alone does not induce acute (7 day) or chronic (28 day) cardiac dysfunction. ICH induces significant neurological and cognitive deficits, as well as acute and chronic cardiac dysfunction compared to sham control mice. Mice subjected to ICH + Spx exhibit significantly improved neurological and cognitive function compared to ICH mice. Mice with ICH + Spx also exhibit significantly improved acute and chronic cardiac function compared to ICH mice indicated by increased left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS), decreased cardiac fibrosis, decreased cardiomyocyte hypertrophy, decreased cardiac infiltration of immune cells and decreased expression of inflammatory factor and oxidative stress in the heart. CONCLUSIONS Our study demonstrates that splenectomy attenuates ICH-induced neurological and cognitive impairment as well as ICH-induced cardiac dysfunction in mice. Inflammatory cell infiltration into heart and immune responses mediated by the spleen may contribute to ICH-induce acute and chronic cardiac dysfunction and pathological cardiac remodeling.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Linlin Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenkui Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA.
| |
Collapse
|
36
|
Guan F, Huang T, Wang X, Xing Q, Gumpper K, Li P, Song J, Tan T, Yang GL, Zang X, Zhang J, Wang Y, Yang Y, Liu Y, Zhang Y, Yang B, Ma J, Ma S. The TRIM protein Mitsugumin 53 enhances survival and therapeutic efficacy of stem cells in murine traumatic brain injury. Stem Cell Res Ther 2019; 10:352. [PMID: 31779687 PMCID: PMC6883632 DOI: 10.1186/s13287-019-1433-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/22/2019] [Accepted: 09/30/2019] [Indexed: 12/13/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a common neurotrauma leading to brain dysfunction and death. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) hold promise in the treatment of TBI. However, their efficacy is modest due to low survival and differentiation under the harsh microenvironment of the injured brain. MG53, a member of TRIM family protein, plays a vital role in cell and tissue damage repair. The present study aims to test whether MG53 preserves hUC-MSCs against oxidative stress and enhances stem cell survival and efficacy in TBI treatment. Methods In this study, we performed a series of in vitro and in vivo experiments in hUC-MSCs and mice to define the function of MG53 enhancing survival, neurogenesis, and therapeutic efficacy of stem cells in murine traumatic brain injury. Results We found that recombinant human MG53 (rhMG53) protein protected hUC-MSCs against H2O2-induced oxidative damage and stimulated hUC-MSC proliferation and migration. In a mouse model of contusion-induced TBI, intravenous administration of MG53 protein preserved the survival of transplanted hUC-MSCs, mitigated brain edema, reduced neurological deficits, and relieved anxiety and depressive-like behaviors. Co-treatment of MG53 and hUC-MSCs enhanced neurogenesis by reducing apoptosis and improving PI3K/Akt-GSK3β signaling. Conclusion MG53 enhances the efficacy of hUC-MSCs in the recovery of TBI, indicating that such adjunctive therapy may provide a novel strategy to lessen damage and optimize recovery for brain injury.
Collapse
Affiliation(s)
- Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.,The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.,Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Tuanjie Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xinxin Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Kristyn Gumpper
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Peng Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jishi Song
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | | | - Xingxing Zang
- Department of Microbiology and Immunology, Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Jiewen Zhang
- Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Yuming Wang
- Henan Provincial People's Hospital, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Yunlei Yang
- Department of Medicine and Neuroscience, Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Yashi Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yanting Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, 43210, USA.
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
37
|
Feng Y, Zheng C, Zhou Z, Xiong H, Feng F, Xie F, Wu ZD. IL-17A neutralizing antibody attenuates eosinophilic meningitis caused by Angiostrongylus cantonensis by involving IL-17RA/Traf6/NF-κB signaling. Exp Cell Res 2019; 384:111554. [DOI: 10.1016/j.yexcr.2019.111554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 01/25/2023]
|
38
|
Han X, Zhao X, Lan X, Li Q, Gao Y, Liu X, Wan J, Yang Z, Chen X, Zang W, Guo AM, Falck JR, Koehler RC, Wang J. 20-HETE synthesis inhibition promotes cerebral protection after intracerebral hemorrhage without inhibiting angiogenesis. J Cereb Blood Flow Metab 2019; 39:1531-1543. [PMID: 29485354 PMCID: PMC6681539 DOI: 10.1177/0271678x18762645] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
20-HETE, an arachidonic acid metabolite synthesized by cytochrome P450 4A, plays an important role in acute brain damage from ischemic stroke or subarachnoid hemorrhage. We tested the hypothesis that 20-HETE inhibition has a protective effect after intracerebral hemorrhage (ICH) and then investigated its effect on angiogenesis. We exposed hippocampal slice cultures to hemoglobin and induced ICH in mouse brains by intrastriatal collagenase injection to investigate the protective effect of 20-HETE synthesis inhibitor N-hydroxy-N'-(4-n-butyl-2-methylphenyl)-formamidine (HET0016). Hemoglobin-induced neuronal death was assessed by propidium iodide after 18 h in vitro. Lesion volume, neurologic deficits, cell death, reactive oxygen species (ROS), neuroinflammation, and angiogenesis were evaluated at different time points after ICH. In cultured mouse hippocampal slices, HET0016 attenuated hemoglobin-induced neuronal death and decreased levels of proinflammatory cytokines and ROS. In vivo, HET0016 reduced brain lesion volume and neurologic deficits, and decreased neuronal death, ROS production, gelatinolytic activity, and the inflammatory response at three days after ICH. However, HET0016 did not inhibit angiogenesis, as levels of CD31, VEGF, and VEGFR2 were unchanged on day 28. We conclude that 20-HETE is involved in ICH-induced brain damage. Inhibition of 20-HETE synthesis may provide a viable means to mitigate ICH injury without inhibition of angiogenesis.
Collapse
Affiliation(s)
- Xiaoning Han
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xiaochun Zhao
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xi Lan
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Qian Li
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yufeng Gao
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xi Liu
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jieru Wan
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zengjin Yang
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xuemei Chen
- 2 Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Weidong Zang
- 2 Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Austin M Guo
- 3 Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | - John R Falck
- 4 Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Raymond C Koehler
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jian Wang
- 1 Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
39
|
Vinukonda G, Liao Y, Hu F, Ivanova L, Purohit D, Finkel DA, Giri P, Bapatla L, Shah S, Zia MT, Hussein K, Cairo MS, La Gamma EF. Human Cord Blood-Derived Unrestricted Somatic Stem Cell Infusion Improves Neurobehavioral Outcome in a Rabbit Model of Intraventricular Hemorrhage. Stem Cells Transl Med 2019; 8:1157-1169. [PMID: 31322326 PMCID: PMC6811700 DOI: 10.1002/sctm.19-0082] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022] Open
Abstract
Intraventricular hemorrhage (IVH) is a severe complication of preterm birth, which leads to hydrocephalus, cerebral palsy, and mental retardation. There are no available therapies to cure IVH, and standard treatment is supportive care. Unrestricted somatic stem cells (USSCs) from human cord blood have reparative effects in animal models of brain and spinal cord injuries. USSCs were administered to premature rabbit pups with IVH and their effects on white matter integrity and neurobehavioral performance were evaluated. USSCs were injected either via intracerebroventricular (ICV) or via intravenous (IV) routes in 3 days premature (term 32d) rabbit pups, 24 hours after glycerol‐induced IVH. The pups were sacrificed at postnatal days 3, 7, and 14 and effects were compared to glycerol‐treated but unaffected or nontreated control. Using in vivo live bioluminescence imaging and immunohistochemical analysis, injected cells were found in the injured parenchyma on day 3 when using the IV route compared to ICV where cells were found adjacent to the ventricle wall forming aggregates; we did not observe any adverse events from either route of administration. The injected USSCs were functionally associated with attenuated microglial infiltration, less apoptotic cell death, fewer reactive astrocytes, and diminished levels of key inflammatory cytokines (TNFα and IL1β). In addition, we observed better preservation of myelin fibers, increased myelin gene expression, and altered reactive astrocyte distribution in treated animals, and this was associated with improved locomotor function. Overall, our findings support the possibility that USSCs exert anti‐inflammatory effects in the injured brain mitigating many detrimental consequences associated with IVH. stem cells translational medicine2019;8:1157–1169
Collapse
Affiliation(s)
- Govindaiah Vinukonda
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,Cell Biology & Anatomy, New York Medical College, Valhalla, New York, USA
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Furong Hu
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Larisa Ivanova
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Deepti Purohit
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Dina A Finkel
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Priyadarshani Giri
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | | | - Shetal Shah
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Muhammed T Zia
- The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Karen Hussein
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,Cell Biology & Anatomy, New York Medical College, Valhalla, New York, USA.,Department of Medicine, Pathology, Microbiology & Immunology, Cell Biology & Anatomy, New York Medical College, Valhalla, New York, USA
| | - Edmund F La Gamma
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA.,The Regional Neonatal Center at Maria Fareri Children's Hospital of Westchester Medical Center, Valhalla, New York, USA.,Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York, USA
| |
Collapse
|
40
|
Zhu H, Wang Z, Yu J, Yang X, He F, Liu Z, Che F, Chen X, Ren H, Hong M, Wang J. Role and mechanisms of cytokines in the secondary brain injury after intracerebral hemorrhage. Prog Neurobiol 2019; 178:101610. [PMID: 30923023 DOI: 10.1016/j.pneurobio.2019.03.003] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 12/18/2022]
Abstract
Intracerebral hemorrhage (ICH) is a common and severe cerebrovascular disease that has high mortality. Few survivors achieve self-care. Currently, patients receive only symptomatic treatment for ICH and benefit poorly from this regimen. Inflammatory cytokines are important participants in secondary injury after ICH. Increases in proinflammatory cytokines may aggravate the tissue injury, whereas increases in anti-inflammatory cytokines might be protective in the ICH brain. Inflammatory cytokines have been studied as therapeutic targets in a variety of acute and chronic brain diseases; however, studies on ICH are limited. This review summarizes the roles and functions of various pro- and anti-inflammatory cytokines in secondary brain injury after ICH and discusses pathogenic mechanisms and emerging therapeutic strategies and directions for treatment of ICH.
Collapse
Affiliation(s)
- Huimin Zhu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhiqiang Wang
- Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Jixu Yu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China; Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiuli Yang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Feng He
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China
| | - Zhenchuan Liu
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Fengyuan Che
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, China; Central laboratory, Linyi People's Hospital, Linyi, Shandong 276003, China.
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, Henan, China
| | - Honglei Ren
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Michael Hong
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
41
|
Microglial Depletion with Clodronate Liposomes Increases Proinflammatory Cytokine Levels, Induces Astrocyte Activation, and Damages Blood Vessel Integrity. Mol Neurobiol 2019; 56:6184-6196. [PMID: 30734229 DOI: 10.1007/s12035-019-1502-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/18/2019] [Indexed: 01/22/2023]
Abstract
Investigators are increasingly interested in using microglial depletion to study the role of microglia under pathologic conditions. Liposome-encapsulated clodronate is commonly used to eliminate macrophage populations because it causes functionally irreversible inhibition and apoptosis once phagocytized by macrophages. Recent studies have shown that microglia can be depleted in disease models by injecting clodronate liposomes into the brain parenchyma. However, it is unclear whether intracerebral administration of clodronate liposomes is a practical method of eliminating microglia under physiologic conditions or whether microglial depletion induces damage to other brain cells. In this study, injecting 1 μL of clodronate liposomes (7 μg/μL) into the striatum of mice caused ablation of microglia at 1 day that persisted for 3 days. Microglia reappeared in the boundary regions of microglia elimination after 5 days. Importantly, we observed an increase in proinflammatory cytokine levels and an increase in neural/glial antigen 2 and glial fibrillary acidic protein expression in the perilesional region. In contrast, expression levels of myelin basic protein, microtubule-associated protein 2, and postsynaptic protein-95 decreased in the periphery of regions where microglia were depleted. Moreover, clodronate liposome administration decreased the density and integrity of blood vessels in the perilesional regions. In cultured primary neurons, clodronate liposome exposure also attenuated ATP synthesis. Together, these findings suggest that intracerebral administration of clodronate liposomes into brain parenchyma can deplete microglia, but can also damage other brain cells and blood vessel integrity.
Collapse
|
42
|
Wang J, Jiang C, Zhang K, Lan X, Chen X, Zang W, Wang Z, Guan F, Zhu C, Yang X, Lu H, Wang J. Melatonin receptor activation provides cerebral protection after traumatic brain injury by mitigating oxidative stress and inflammation via the Nrf2 signaling pathway. Free Radic Biol Med 2019; 131:345-355. [PMID: 30553970 DOI: 10.1016/j.freeradbiomed.2018.12.014] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/09/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023]
Abstract
Traumatic brain injury (TBI) is a principal cause of death and disability worldwide. Melatonin, a hormone made by the pineal gland, is known to have anti-inflammatory and antioxidant properties. In this study, using a weight-drop model of TBI, we investigated the protective effects of ramelteon, a melatonin MT1/MT2 receptor agonist, and its underlying mechanisms of action. Administration of ramelteon (10 mg/kg) daily at 10:00 a.m. alleviated TBI-induced early brain damage on day 3 and long-term neurobehavioral deficits on day 28 in C57BL/6 mice. Ramelteon also increased the protein levels of interleukin (IL)-10, IL-4, superoxide dismutase (SOD), glutathione, and glutathione peroxidase and reduced the protein levels of IL-1β, tumor necrosis factor, and malondialdehyde in brain tissue and serum on days 1, 3, and 7 post-TBI. Similarly, ramelteon attenuated microglial and astrocyte activation in the perilesional cortex on day 3. Furthermore, ramelteon decreased Keap 1 expression, promoted nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation, and increased levels of downstream proteins, including SOD-1, heme oxygenase-1, and NQO1 on day 3 post-TBI. However, in Nrf2 knockout mice with TBI, ramelteon did not decrease the lesion volume, neuronal degeneration, or myelin loss on day 3; nor did it mitigate depression-like behavior or most motor behavior deficits on day 28. Thus, timed ramelteon treatment appears to prevent inflammation and oxidative stress via the Nrf2-antioxidant response element pathway and might represent a potential chronotherapeutic strategy for treating TBI.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Antioxidants/pharmacology
- Astrocytes/drug effects
- Astrocytes/metabolism
- Astrocytes/pathology
- Brain Edema/drug therapy
- Brain Edema/genetics
- Brain Edema/metabolism
- Brain Edema/pathology
- Brain Injuries, Traumatic/drug therapy
- Brain Injuries, Traumatic/genetics
- Brain Injuries, Traumatic/metabolism
- Brain Injuries, Traumatic/pathology
- Cerebral Cortex/drug effects
- Cerebral Cortex/metabolism
- Cerebral Cortex/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Glutathione Peroxidase/genetics
- Glutathione Peroxidase/metabolism
- Indenes/pharmacology
- Inflammation
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Interleukin-1beta/genetics
- Interleukin-1beta/metabolism
- Interleukin-4/genetics
- Interleukin-4/metabolism
- Kelch-Like ECH-Associated Protein 1/genetics
- Kelch-Like ECH-Associated Protein 1/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/drug effects
- Microglia/metabolism
- Microglia/pathology
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Oxidative Stress/drug effects
- Receptor, Melatonin, MT1/agonists
- Receptor, Melatonin, MT1/genetics
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT2/agonists
- Receptor, Melatonin, MT2/genetics
- Receptor, Melatonin, MT2/metabolism
- Signal Transduction
- Superoxide Dismutase/genetics
- Superoxide Dismutase/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Junmin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China; The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Weidong Zang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Zhongyu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Fangxia Guan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; School of Life Science, Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Changlian Zhu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan 450052, China; Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Göteborg 40530, Sweden
| | - Xiuli Yang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
43
|
Li Q, Lan X, Han X, Wang J. Expression of Tmem119/ Sall1 and Ccr2/ CD69 in FACS-Sorted Microglia- and Monocyte/Macrophage-Enriched Cell Populations After Intracerebral Hemorrhage. Front Cell Neurosci 2019; 12:520. [PMID: 30687011 PMCID: PMC6333739 DOI: 10.3389/fncel.2018.00520] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/12/2018] [Indexed: 11/30/2022] Open
Abstract
Activation and polarization of microglia and macrophages are critical events in neuroinflammation and hematoma resolution after intracerebral hemorrhage (ICH). However, distinguishing microglia and monocyte-derived macrophages histologically can be difficult. Although they share most cell surface markers, evidence indicates that the gene regulation and function of these two cell types might be different. Flow cytometry is the gold standard for discriminating between the two cell populations, but it is rarely used in the ICH research field. We developed a flow cytometry protocol to identify and sort microglia and monocyte-derived macrophages from mice that have undergone well-established ICH models induced by collagenase or blood injection. In addition, we combined a recently established magnetic-activated cell separation system that allows eight tissue samples to be assessed together. This protocol can be completed within 5–8 h. Sorted cells are fully preserved and maintain expression of microglia-specific (Tmem119/Sall1) and macrophage-specific (Ccr2/CD69) markers. They retain phagocytic ability, respond to lipopolysaccharide stimulation, and engulf fluorescent latex beads. Thus, this protocol represents a very important tool for researching microglial and monocyte-derived macrophage biologic function after ICH and other brain diseases.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Li W, Li L, Chopp M, Venkat P, Zacharek A, Chen Z, Landschoot-Ward J, Yan T, Chen J. Intracerebral Hemorrhage Induces Cardiac Dysfunction in Mice Without Primary Cardiac Disease. Front Neurol 2018; 9:965. [PMID: 30524357 PMCID: PMC6256175 DOI: 10.3389/fneur.2018.00965] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Intracerebral hemorrhage (ICH) is a life threatening stroke subtype and a worldwide health problem. In this study, we investigate brain-heart interaction after ICH in mice and test whether ICH induces cardiac dysfunction in the absence of primary cardiac disease. We also investigate underlying mechanisms such as oxidative stress and inflammatory responses in mediating cardiac dysfunction post-ICH in mice. Methods: Male, adult (3–4 m) C57BL/6J mice were subjected to sham surgery or ICH using an autologous blood injection model (n = 16/group). Cardiac function was evaluated at 7 and 28 days after ICH using echocardiography (n = 8/group per time point). Western blot and immunostaining analysis were employed to assess oxidative stress and inflammatory responses in the heart. Results: Mice subjected to ICH exhibited significantly decreased cardiac contractile function measured by left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) at 7 and 28 days after ICH compared to sham-control mice (p < 0.05). ICH induced cardiac dysfunction was significantly worse at 28 days than at 7 days after ICH (p < 0.05). ICH in mice significantly increased cardiomyocyte apoptosis, inflammatory factor expression and inflammatory cell infiltration in heart tissue, and induced cardiac oxidative stress at 7 days post-ICH compared to sham-control mice. Compared to sham-control mice, ICH-mice also exhibited significantly increased (p < 0.05) cardiomyocyte hypertrophy and cardiac fibrosis at 28 days after ICH. Conclusions: ICH induces significant and progressive cardiac dysfunction in mice. ICH increases cardiac oxidative stress and inflammatory factor expression in heart tissue which may play key roles in ICH-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Wei Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Neurology, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in CNS, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Linlin Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Neurology, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in CNS, Ministry of Education and Tianjin City, Tianjin, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, NY, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Zhili Chen
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Neurology, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in CNS, Ministry of Education and Tianjin City, Tianjin, China.,Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Tao Yan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Neurology, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in CNS, Ministry of Education and Tianjin City, Tianjin, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
45
|
Vafaee F, Zarifkar A, Emamghoreishi M, Namavar MR, Shahpari M, Zarifkar AH. Effect of Recombinant Insulin-like Growth Factor-2 Injected into the Hippocampus on Memory Impairment Following Hippocampal Intracerebral Hemorrhage in Rats. Galen Med J 2018; 7:e1353. [PMID: 34466449 PMCID: PMC8344085 DOI: 10.22086/gmj.v0i0.1353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 09/28/2018] [Accepted: 10/25/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Insulin-like growth factor 2 (IGF-2) is a growth factor and an anti-inflammatory cytokine that plays a pivotal role in memory. In this study, we examined the effect of recombinant IGF-2 on memory impairment due to intracerebral hemorrhage (ICH). Avoidance and recognition memory, locomotor activity, neurological deficit score (NDS), and the level of the IGF-2 gene expression were evaluated. MATERIALS AND METHODS To induce ICH, 100 μL of autologous blood was injected into the left hippocampus of male Sprague Dawley rats. Recombinant IGF-2 was injected into the damaged hippocampus 30 minutes after the induction of ICH. Then, over two weeks, NDS, locomotor activity, passive avoidance, and novel object recognition (NOR) test were evaluated. Finally, the level of IGF-2 gene expression was evaluated by using the real-time polymerase chain reaction technique. RESULT Our results indicated that recombinant IGF-2 injection significantly increased step-through latency (P<0.001) and total time spent in the dark box (P<0.01). However, no significant difference was seen in recognition memory and NDS. Locomotor activity did not significantly change in any group. A significantly reduced level of IGF-2 was observed after two weeks (P<0.05). CONCLUSION The results of this study show that a single dose of recombinant IGF-2 injection can influence hippocampus-dependent memories. Importantly, IGF-2 did not change locomotor activity and NDS after two weeks, which probably represents its specific function in memory.
Collapse
Affiliation(s)
- Farzaneh Vafaee
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Emamghoreishi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Shahpari
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Hossein Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
46
|
Li Q, Weiland A, Chen X, Lan X, Han X, Durham F, Liu X, Wan J, Ziai WC, Hanley DF, Wang J. Ultrastructural Characteristics of Neuronal Death and White Matter Injury in Mouse Brain Tissues After Intracerebral Hemorrhage: Coexistence of Ferroptosis, Autophagy, and Necrosis. Front Neurol 2018; 9:581. [PMID: 30065697 PMCID: PMC6056664 DOI: 10.3389/fneur.2018.00581] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/27/2018] [Indexed: 11/24/2022] Open
Abstract
Although intracerebral hemorrhage (ICH) is a devastating disease worldwide, the pathologic changes in ultrastructure during the acute and chronic phases of ICH are poorly described. In this study, transmission electron microscopy was used to examine the ultrastructure of ICH-induced pathology. ICH was induced in mice by an intrastriatal injection of collagenase. Pathologic changes were observed in the acute (3 days), subacute (6 days), and chronic (28 days) phases. Compared with sham animals, we observed various types of cell death in the injured striatum during the acute phase of ICH, including necrosis, ferroptosis, and autophagy. Different degrees of axon degeneration in the striatum were seen in the acute phase, and axonal demyelination was observed in the ipsilateral striatum and corpus callosum at late time points. In addition, phagocytes, resident microglia, and infiltrating monocyte-macrophages were present around red blood cells and degenerating neurons and were observed to engulf red blood cells and other debris. Many synapses appeared abnormal or were lost. This systematic analysis of the pathologic changes in ultrastructure after ICH in mice provides information that will be valuable for future ICH pathology studies.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, Beijing, China
| | - Abigail Weiland
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Frederick Durham
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xi Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wendy C. Ziai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel F. Hanley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
47
|
Human Placenta-Derived Mesenchymal Stem Cells Reduce Mortality and Hematoma Size in a Rat Intracerebral Hemorrhage Model in an Acute Phase. Stem Cells Int 2018; 2018:1658195. [PMID: 29853907 PMCID: PMC5954892 DOI: 10.1155/2018/1658195] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/05/2018] [Accepted: 04/08/2018] [Indexed: 01/01/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a critical disease, highly associated with mortality and morbidity. Several studies have demonstrated the beneficial effect of mesenchymal stem cells (MSCs) on ICH, mostly focused on their mid-to-long-term effect. Acute hematoma expansion is one of the most important prognostic factors of ICH. We hypothesized that MSCs would decrease mortality and hematoma size in acute ICH, based on the findings of a few recent researches reporting their effect on blood-brain barrier and endothelial integrity. Rat ICH models were made using bacterial collagenase. One hour after ICH induction, the rats were randomly divided into MSC-treated and control groups. Mortality, hematoma volume, ventricular enlargement, brain edema, and degenerating neuron count were compared at 24 hours after ICH induction. Expression of tight junction proteins (ZO-1, occludin) and coagulation factor VII mRNA was also compared. Mortality rate (50% versus 8.3%), hematoma size, ventricular size, hemispheric enlargement, and degenerating neuron count were significantly lower in the MSC-treated group (p = 0.034, 0.038, 0.001, 0.022, and <0.001, resp.), while the expression of ZO-1 and occludin was higher (p = 0.007 and 0.012). Administration of MSCs may prevent hematoma expansion in the hyperacute stage of ICH and decrease acute mortality by enhancing the endothelial integrity of cerebral vasculature.
Collapse
|
48
|
Akhter M, Qin T, Fischer P, Sadeghian H, Kim HH, Whalen MJ, Goldstein JN, Ayata C. Rho-kinase inhibitors do not expand hematoma volume in acute experimental intracerebral hemorrhage. Ann Clin Transl Neurol 2018; 5:769-776. [PMID: 29928660 PMCID: PMC5989779 DOI: 10.1002/acn3.569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 01/13/2023] Open
Abstract
Rho‐associated kinase (ROCK) is an emerging target in acute ischemic stroke. Early pre‐hospital treatment with ROCK inhibitors may improve their efficacy, but their antithrombotic effects raise safety concerns in hemorrhagic stroke, precluding use prior to neuroimaging. Therefore, we tested whether ROCK inhibition affects the bleeding times, and worsens hematoma volume in a model of intracerebral hemorrhage (ICH) induced by intrastriatal collagenase injection in mice. Tail bleeding time was measured 1 h after treatment with isoform‐nonselective inhibitor fasudil, or ROCK2‐selective inhibitor KD025, or their vehicles. In the ICH model, treatments were administered 1 h after collagenase injection. Although KD025 but not fasudil prolonged the tail bleeding times, neither drug expanded the volume of ICH or worsened neurological deficits at 48 h compared with vehicle. Although more testing is needed in aged animals and comorbid models such as diabetes, these results suggest ROCK inhibitors may be safe for pre‐hospital administration in acute stroke.
Collapse
Affiliation(s)
- Murtaza Akhter
- Department of Emergency Medicine University of Arizona College of Medicine- Phoenix Maricopa Medical Center Phoenix Arizona.,Neurovascular Research Laboratory Department of Radiology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts.,Department of Emergency Medicine Massachusetts General Hospital Harvard Medical School Boston Massachusetts
| | - Tom Qin
- Neurovascular Research Laboratory Department of Radiology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts
| | - Paul Fischer
- Neurovascular Research Laboratory Department of Radiology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts
| | - Homa Sadeghian
- Neurovascular Research Laboratory Department of Radiology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts
| | - Hyung Hwan Kim
- Neurovascular Research Laboratory Department of Radiology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts
| | - Michael J Whalen
- Department of Pediatrics Pediatric Critical Care Medicine Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts
| | - Joshua N Goldstein
- Department of Emergency Medicine Massachusetts General Hospital Harvard Medical School Boston Massachusetts
| | - Cenk Ayata
- Neurovascular Research Laboratory Department of Radiology Massachusetts General Hospital Harvard Medical School Charlestown Massachusetts.,Stroke Service and Neuroscience Intensive Care Unit Department of Neurology Massachusetts General Hospital Harvard Medical School Boston Massachusetts
| |
Collapse
|
49
|
Koschnitzky JE, Keep RF, Limbrick DD, McAllister JP, Morris JA, Strahle J, Yung YC. Opportunities in posthemorrhagic hydrocephalus research: outcomes of the Hydrocephalus Association Posthemorrhagic Hydrocephalus Workshop. Fluids Barriers CNS 2018; 15:11. [PMID: 29587767 PMCID: PMC5870202 DOI: 10.1186/s12987-018-0096-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/09/2018] [Indexed: 12/19/2022] Open
Abstract
The Hydrocephalus Association Posthemorrhagic Hydrocephalus Workshop was held on July 25 and 26, 2016 at the National Institutes of Health. The workshop brought together a diverse group of researchers including pediatric neurosurgeons, neurologists, and neuropsychologists with scientists in the fields of brain injury and development, cerebrospinal and interstitial fluid dynamics, and the blood-brain and blood-CSF barriers. The goals of the workshop were to identify areas of opportunity in posthemorrhagic hydrocephalus research and encourage scientific collaboration across a diverse set of fields. This report details the major themes discussed during the workshop and research opportunities identified for posthemorrhagic hydrocephalus. The primary areas include (1) preventing intraventricular hemorrhage, (2) stopping primary and secondary brain damage, (3) preventing hydrocephalus, (4) repairing brain damage, and (5) improving neurodevelopment outcomes in posthemorrhagic hydrocephalus.
Collapse
Affiliation(s)
| | - Richard F. Keep
- University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109 USA
| | - David D. Limbrick
- Washington University in St. Louis School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - James P. McAllister
- Washington University in St. Louis School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - Jill A. Morris
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Neuroscience Center, 6001 Executive Blvd, NSC Rm 2112, Bethesda, MD 20892 USA
| | - Jennifer Strahle
- Washington University in St. Louis School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110 USA
| | - Yun C. Yung
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd., Building 7, La Jolla, CA 92037 USA
| |
Collapse
|
50
|
Zhu W, Gao Y, Wan J, Lan X, Han X, Zhu S, Zang W, Chen X, Ziai W, Hanley DF, Russo SJ, Jorge RE, Wang J. Changes in motor function, cognition, and emotion-related behavior after right hemispheric intracerebral hemorrhage in various brain regions of mouse. Brain Behav Immun 2018; 69:568-581. [PMID: 29458197 PMCID: PMC5857479 DOI: 10.1016/j.bbi.2018.02.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/25/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a detrimental type of stroke. Mouse models of ICH, induced by collagenase or blood infusion, commonly target striatum, but not other brain sites such as ventricular system, cortex, and hippocampus. Few studies have systemically investigated brain damage and neurobehavioral deficits that develop in animal models of ICH in these areas of the right hemisphere. Therefore, we evaluated the brain damage and neurobehavioral dysfunction associated with right hemispheric ICH in ventricle, cortex, hippocampus, and striatum. The ICH model was induced by autologous whole blood or collagenase VII-S (0.075 units in 0.5 µl saline) injection. At different time points after ICH induction, mice were assessed for brain tissue damage and neurobehavioral deficits. Sham control mice were used for comparison. We found that ICH location influenced features of brain damage, microglia/macrophage activation, and behavioral deficits. Furthermore, the 24-point neurologic deficit scoring system was most sensitive for evaluating locomotor abnormalities in all four models, especially on days 1, 3, and 7 post-ICH. The wire-hanging test was useful for evaluating locomotor abnormalities in models of striatal, intraventricular, and cortical ICH. The cylinder test identified locomotor abnormalities only in the striatal ICH model. The novel object recognition test was effective for evaluating recognition memory dysfunction in all models except for striatal ICH. The tail suspension test, forced swim test, and sucrose preference test were effective for evaluating emotional abnormality in all four models but did not correlate with severity of brain damage. These results will help to inform future preclinical studies of ICH outcomes.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Yufeng Gao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jieru Wan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shanshan Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Weidong Zang
- Department of Human Anatomy, Basic Medical College of Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Xuemei Chen
- Department of Human Anatomy, Basic Medical College of Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Wendy Ziai
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel F Hanley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo E Jorge
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Human Anatomy, Basic Medical College of Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|