1
|
Xie Q, Liao YH, He WJ, Wang GQ. Incidence and clinical analysis of asymptomatic intracranial hemorrhage in neonates with cerebral hypoxic-ischaemic risk based on multisequence MR images. Sci Rep 2024; 14:14721. [PMID: 38926428 PMCID: PMC11208507 DOI: 10.1038/s41598-024-62473-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
The incidence and clinical distribution of intracranial haemorrhage (ICH) in neonates at risk of cerebral hypoxia-ischaemia have not been reported in specific studies. Based on conventional magnetic resonance imaging (MRI) versus susceptibility weighted imaging (SWI), this study aimed to analyse the occurrence of asymptomatic ICH in newborns with or without risk of cerebral hypoxia-ischaemia and to accumulate objective data for clinical evaluations of high-risk neonates and corresponding response strategies. 317 newborns were included. MRI revealed that the overall incidence of ICH was 59.31%. The most common subtype was intracranial extracerebral haemorrhage (ICECH) which included subarachnoid haemorrhage (SAH) and subdural haemorrhage (SDH). ICECH accounted for 92.02% of ICH. The positive detection rate of ICECH by SWI was significantly higher than that by T1WI. The incidence of total ICH, ICECH and SAH was greater among children who were delivered vaginally than among those who underwent caesarean delivery. Asymptomatic neonatal ICH may be a common complication of the neonatal birth process, and SWI may improve the detection rate. Transvaginal delivery and a weight greater than 2500 g were associated with a high incidence of ICECH in neonates. The impact of neonatal cerebral hypoxia-ischaemia risk factors on the occurrence of asymptomatic ICH may be negligible.
Collapse
Affiliation(s)
- Qi Xie
- Department of Medical Imaging in Nansha, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 511457, China.
| | - Yan-Hui Liao
- Department of Medical Imaging in Nansha, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 511457, China
- Department of Nuclear Medicine, Meizhou People's Hospital, Meizhou, 514031, Guangdong, China
| | - Wen-Juan He
- Department of Medical Imaging in Nansha, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 511457, China
| | - Gui-Qin Wang
- Medical Record Department in Nansha, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 511457, China
| |
Collapse
|
2
|
Cao Y, Yao X. Acute albumin administration as therapy for intracerebral hemorrhage: A literature review. Heliyon 2024; 10:e23946. [PMID: 38192834 PMCID: PMC10772721 DOI: 10.1016/j.heliyon.2023.e23946] [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: 06/14/2023] [Revised: 11/05/2023] [Accepted: 12/16/2023] [Indexed: 01/10/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke with high mortality. Secondary brain injury after surviving the initial ictus leads to severe neurological deficits, and has emerged as an attractive therapeutic target. Human serum albumin (HSA), a pluripotent protein synthesized mainly in the liver, has shown remarkable efficacy by targeting secondary brain injury pathways in rodent models of ICH, while results from relevant clinical research on albumin therapy remain unclear. Preclinical studies have shown albumin-mediated neuroprotection may stem from its biological functions, including its major antioxidation activity, anti-inflammatory responses, and anti-apoptosis. HSA treatment provides neuroprotective and recovery enhancement effects via improving short and long-term neurologic function, maintaining blood-brain barrier (BBB) integrity and reducing neuronal oxidative stress and apoptosis. Retrospective clinical studies have shown that admission hypoalbuminemia is a prognostic factor for poor outcomes in patients with ICH. However, clinical trial was terminated due to poor enrollment and its potential adverse effects. This review provides an overview of the physiological properties of albumin, as well as its potential neuroprotective and prognostic value and the resulting clinical implications.
Collapse
Affiliation(s)
- Yirong Cao
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiaoying Yao
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
3
|
Sawaya R, Ueda J, Saito S. Quantitative Susceptibility Mapping and Amide Proton Transfer-Chemical Exchange Saturation Transfer for the Evaluation of Intracerebral Hemorrhage Model. Int J Mol Sci 2023; 24:ijms24076627. [PMID: 37047596 PMCID: PMC10095413 DOI: 10.3390/ijms24076627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/23/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
This study aimed to evaluate an intracerebral hemorrhage (ICH) model using quantitative susceptibility mapping (QSM) and chemical exchange saturation transfer (CEST) with preclinical 7T-magnetic resonance imaging (MRI) and determine the potential of amide proton transfer-CEST (APT-CEST) for use as a biomarker for the early detection of ICH. Six Wistar male rats underwent MRI, and another six underwent histopathological examinations on postoperative days 0, 3, and 7. The ICH model was created by injecting bacterial collagenase into the right hemisphere of the brain. QSM and APT-CEST MRI were performed using horizontal 7T-MRI. Histological studies were performed to observe ICH and detect iron deposition at the ICH site. T2-weighted images (T2WI) revealed signal changes associated with hemoglobin degeneration in red blood cells, indicating acute-phase hemorrhage on day 0, late-subacute-phase hemorrhage on day 3, and chronic-phase hemorrhage on day 7. The susceptibility alterations in each phase were detected using QSM. QSM and Berlin blue staining revealed hemosiderin deposition in the chronic phase. APT-CEST revealed high magnetization transfer ratios in the acute phase. Abundant mobile proteins and peptides were observed in early ICH, which were subsequently diluted. APT-CEST imaging may be a reliable noninvasive biomarker for the early diagnosis of ICH.
Collapse
Affiliation(s)
- Reika Sawaya
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita 560-0871, Osaka, Japan
| | - Junpei Ueda
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita 560-0871, Osaka, Japan
| | - Shigeyoshi Saito
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita 560-0871, Osaka, Japan
- Department of Advanced Medical Technologies, National Cardiovascular and Cerebral Research Center, Suita 564-8565, Osaka, Japan
| |
Collapse
|
4
|
Boudreau E, Kerwin SC, DuPont EB, Levine JM, Griffin JF. Temporal and sequence-related variability in diffusion-weighted imaging of presumed cerebrovascular accidents in the dog brain. Front Vet Sci 2022; 9:1008447. [PMID: 36419725 PMCID: PMC9676236 DOI: 10.3389/fvets.2022.1008447] [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: 07/31/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
Diffusion-weighted MRI (DWI) is often used to guide clinical interpretation of intraparenchymal brain lesions when there is suspicion for a cerebrovascular accident (CVA). Despite widespread evidence that imaging and patient parameters can influence diffusion-weighted measurements, such as apparent diffusion coefficient (ADC), there is little published data on such measurements for naturally occurring CVA in clinical cases in dogs. We describe a series of 22 presumed and confirmed spontaneous canine CVA with known time of clinical onset imaged on a single 3T magnet between 2011 and 2021. Median ADC values of < 1.0x10−3 mm2/s were seen in normal control tissues as well as within CVAs. Absolute and relative ADC values in CVAs were well-correlated (R2 = 0.82). Absolute ADC values < 1.0x10−3 mm2/s prevailed within ischemic CVAs, though there were exceptions, including some lesions of < 5 days age. Some lesions showed reduced absolute but not relative ADC values when compared to matched normal contralateral tissue. CVAs with large hemorrhagic components did not show restricted diffusion. Variation in the DWI sequence used impacted the ADC values obtained. Failure to identify a region of ADC < 1.0x10−3 mm2/s should not exclude CVA from the differential list when clinical suspicion is high.
Collapse
Affiliation(s)
- Elizabeth Boudreau
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
- *Correspondence: Elizabeth Boudreau
| | - Sharon C. Kerwin
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Emily B. DuPont
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Jonathan M. Levine
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - John F. Griffin
- Department of Large Animal Clinical Sciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
5
|
Siddiqui EM, Mehan S, Bhalla S, Shandilya A. Potential role of IGF-1/GLP-1 signaling activation in intracerebral hemorrhage. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100055. [PMID: 36685765 PMCID: PMC9846475 DOI: 10.1016/j.crneur.2022.100055] [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: 12/12/2021] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 01/25/2023] Open
Abstract
IGF-1 and GLP-1 receptors are essential in all tissues, facilitating defense by upregulating anabolic processes. They are abundantly distributed throughout the central nervous system, promoting neuronal proliferation, survival, and differentiation. IGF-1/GLP-1 is a growth factor that stimulates neurons' development, reorganization, myelination, and survival. In primary and secondary brain injury, the IGF-1/GLP-1 receptors are impaired, resulting in further neuro complications such as cerebral tissue degradation, neuroinflammation, oxidative stress, and atrophy. Intracerebral hemorrhage (ICH) is a severe condition caused by a stroke for which there is currently no effective treatment. While some pre-clinical studies and medications are being developed as symptomatic therapies in clinical trials, there are specific pharmacological implications for improving post-operative conditions in patients with intensive treatment. Identifying the underlying molecular process and recognizing the worsening situation can assist researchers in developing effective therapeutic solutions to prevent post-hemorrhagic symptoms and the associated neural dysfunctions. As a result, in the current review, we have addressed the manifestations of the disease that are aggravated by the downregulation of IGF-1 and GLP-1 receptors, which can lead to ICH or other neurodegenerative disorders. Our review summarizes that IGF-1/GLP-1 activators may be useful for treating ICH and its related neurodegeneration.
Collapse
Affiliation(s)
- Ehraz Mehmood Siddiqui
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sonalika Bhalla
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Ambika Shandilya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
6
|
Zhang R, Yong VW, Xue M. Revisiting Minocycline in Intracerebral Hemorrhage: Mechanisms and Clinical Translation. Front Immunol 2022; 13:844163. [PMID: 35401553 PMCID: PMC8993500 DOI: 10.3389/fimmu.2022.844163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/24/2022] [Indexed: 01/31/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is an important subtype of stroke with an unsatisfactory prognosis of high mortality and disability. Although many pre-clinical studies and clinical trials have been performed in the past decades, effective therapy that meaningfully improve prognosis and outcomes of ICH patients is still lacking. An active area of research is towards alleviating secondary brain injury after ICH through neuroprotective pharmaceuticals and in which minocycline is a promising candidate. Here, we will first discuss new insights into the protective mechanisms of minocycline for ICH including reducing iron-related toxicity, maintenance of blood-brain barrier, and alleviating different types of cell death from preclinical data, then consider its shortcomings. Finally, we will review clinical trial perspectives for minocycline in ICH. We hope that this summary and discussion about updated information on minocycline as a viable treatment for ICH can facilitate further investigations.
Collapse
Affiliation(s)
- Ruiyi Zhang
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - V. Wee Yong
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Mengzhou Xue
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Longitudinal Observation of Asymmetric Iron Deposition in an Intracerebral Hemorrhage Model Using Quantitative Susceptibility Mapping. Symmetry (Basel) 2022. [DOI: 10.3390/sym14020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Quantitative susceptibility mapping (QSM) is used to obtain quantitative magnetic susceptibility maps of materials from magnitude and phase images acquired by three-dimensional gradient-echo using inverse problem-solving. Few preclinical studies have evaluated the intracerebral hemorrhage (ICH) model and asymmetric iron deposition. We created a rat model of ICH and compared QSM and conventional magnetic resonance imaging (MRI) during the longitudinal evaluation of ICH. Collagenase was injected in the right striatum of 12-week-old Wistar rats. QSM and conventional MRI were performed on days 0, 1, 7, and 28 after surgery using 7-Tesla MRI. Susceptibility, normalized signal value, and area of the hemorrhage site were statistically compared during image analysis. Susceptibility decreased monotonically up to day 7 but increased on day 28. Other imaging methods showed a significant increase in signal from day 0 to day 1 but a decreasing trend after day 1. During the area evaluation, conventional MRI methods showed an increase from day 0 to day 1; however, decreases were observed thereafter. QSM showed a significant increase from day 0 to day 1. The temporal evaluation of ICH by QSM suggested the possibility of detecting of asymmetric iron deposition for normal brain site.
Collapse
|
8
|
Gómez-de Frutos MC, García-Suárez I, Laso-García F, Diekhorst L, Otero-Ortega L, Alonso de Leciñana M, Fuentes B, Gutiérrez-Fernández M, Díez-Tejedor E, Ruíz-Ares G. B-Mode Ultrasound, a Reliable Tool for Monitoring Experimental Intracerebral Hemorrhage. Front Neurol 2022; 12:771402. [PMID: 35002926 PMCID: PMC8733327 DOI: 10.3389/fneur.2021.771402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Magnetic resonance imaging (MRI) is currently used for the study of intracerebral hemorrhage (ICH) in animal models. However, ultrasound is an inexpensive, non-invasive and rapid technique that could facilitate the diagnosis and follow-up of ICH. This study aimed to evaluate the feasibility and reliability of B-mode ultrasound as an alternative tool for in vivo monitoring of ICH volume and brain structure displacement in an animal model. Methods: A total of 31 male and female Sprague-Dawley rats were subjected to an ICH model using collagenase-IV in the striatum following stereotaxic references. The animals were randomly allocated into 3 groups: healthy (n = 10), sham (n = 10) and ICH (n = 11). B-mode ultrasound studies with a 13-MHz probe were performed pre-ICH and at 5 h, 48 h, 4 d and 1 mo post-ICH for the assessment of ICH volume and displacement of brain structures, considering the distance between the subarachnoid cisterns and the dura mater. The same variables were studied by MRI at 48 h and 1 mo post-ICH. Results: Both imaging techniques showed excellent correlation in measuring ICH volume at 48 h (r = 0.905) and good at 1 mo (r = 0.656). An excellent correlation was also observed in the measured distance between the subarachnoid cisterns and the dura mater at 1 mo between B-mode ultrasound and MRI, on both the ipsilateral (r = 0.870) and contralateral (r = 0.906) sides of the lesion. Conclusion: B-mode ultrasound imaging appears to be a reliable tool for in vivo assessment of ICH volume and displacement of brain structures in animal models.
Collapse
Affiliation(s)
- Mari Carmen Gómez-de Frutos
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Iván García-Suárez
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain.,Department of Emergency Service, San Agustín Hospital, University of San Agustin, Asturias, Spain
| | - Fernando Laso-García
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Luke Diekhorst
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Alonso de Leciñana
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Blanca Fuentes
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Gerardo Ruíz-Ares
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, Neuroscience Area of IdiPAZ Health Research Institute, La Paz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
9
|
Neuroprotective Therapies for Spontaneous Intracerebral Hemorrhage. Neurocrit Care 2021; 35:862-886. [PMID: 34341912 DOI: 10.1007/s12028-021-01311-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022]
Abstract
Patients who survive the initial ictus of spontaneous intracerebral hemorrhage (ICH) remain vulnerable to subsequent injury of the perilesional parenchyma by molecular and cellular responses to the hematoma. Secondary brain injury after ICH, which contributes to long-term functional impairment and mortality, has emerged as an attractive therapeutic target. This review summarizes preclinical and clinical evidence for neuroprotective therapies targeting secondary injury pathways following ICH. A focus on therapies with pleiotropic antiinflammatory effects that target thrombin-mediated chemotaxis and inflammatory cell migration has led to studies investigating statins, anticholinergics, sphingosine-1-phosphate receptor modulators, peroxisome proliferator activated receptor gamma agonists, and magnesium. Attempts to modulate ICH-induced blood-brain barrier breakdown and perihematomal edema formation has prompted studies of nonsteroidal antiinflammatory agents, matrix metalloproteinase inhibitors, and complement inhibitors. Iron chelators, such as deferoxamine and albumin, have been used to reduce the free radical injury that ensues from erythrocyte lysis. Stem cell transplantation has been assessed for its potential to enhance subacute neurogenesis and functional recovery. Despite promising preclinical results of numerous agents, their outcomes have not yet translated into positive clinical trials in patients with ICH. Further studies are necessary to improve our understanding of the molecular events that promote damage and inflammation of the perihematomal parenchyma after ICH. Elucidating the temporal and pathophysiologic features of this secondary brain injury could enhance the clinical efficacy of neuroprotective therapies for ICH.
Collapse
|
10
|
Ren S, Wu G, Huang Y, Wang L, Li Y, Zhang Y. MiR-18a Aggravates Intracranial Hemorrhage by Regulating RUNX1-Occludin/ZO-1 Axis to Increase BBB Permeability. J Stroke Cerebrovasc Dis 2021; 30:105878. [PMID: 34077824 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105878] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 04/27/2021] [Accepted: 05/05/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES To study the molecular mechanisms of miR-18a aggravating intracranial hemorrhage (ICH) by increasing the blood-brain barrier (BBB) permeability. METHODS Brain microvascular endothelial cells (BMVECs) and astrocytes were isolated, identified, and co-cultured to establish in vitro BBB model. BMVECs co-cultured with astrocytes were stimulated with or without thrombase and then transfected with miR-18a mimic and/or si-RUNX1. The trans-endothelial electric resistance (TEER) and FlNa flux were measured, respectively. The potential interaction between RUNX1 and miR-18a was also detected. Additionally, SD rats were injected with fresh autologous non-anticoagulant blood into the brain basal ganglia to establish ICH model. After administration with miR-18a, sh-miR-18a, miR-18a+RUNX1, sh-miR-18a+sh-RUNX1, respectively, BBB permeability was assessed. RESULTS After overexpressing miR-18a, the expression levels of RUNX1, Occludin and ZO-1 were decreased, but the Evan's blue contents and brain water contents were significantly increased in ICH rats. Additionally, rat neurological function was impaired, accompanying with an increase of TEER and fluorescein sodium flux. MiR-18a was a direct target of RUNX1 and it could bind to the promoters of RUNX1 to inhibit the expression of Occuldin and ZO-1. Consistently, these phenomena could also be observed in the corresponding cell model. Conversely, miR-18a knockdown or RUNX1 overexpression just presented an improvement effect on ICH. CONCLUSIONS MiR-18a plays a critical role during ICH because it targets to RUNX1 to inhibit the expression of tight junction proteins (Occludin and ZO-1) and then disrupt BBB permeability. MiR-18a might be a probable therapeutic target for ICH diseases.
Collapse
Affiliation(s)
- Siying Ren
- Guizhou Medical University, Guiyang 550025, China; Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Guofeng Wu
- Guizhou Medical University, Guiyang 550025, China; Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.
| | - Yuanxin Huang
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Likun Wang
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Yinghui Li
- Guizhou Medical University, Guiyang 550025, China
| | - Yan Zhang
- Guizhou Medical University, Guiyang 550025, China; Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
11
|
Van Avondt K, Nur E, Zeerleder S. Mechanisms of haemolysis-induced kidney injury. Nat Rev Nephrol 2019; 15:671-692. [PMID: 31455889 DOI: 10.1038/s41581-019-0181-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
Intravascular haemolysis is a fundamental feature of chronic hereditary and acquired haemolytic anaemias, including those associated with haemoglobinopathies, complement disorders and infectious diseases such as malaria. Destabilization of red blood cells (RBCs) within the vasculature results in systemic inflammation, vasomotor dysfunction, thrombophilia and proliferative vasculopathy. The haemoprotein scavengers haptoglobin and haemopexin act to limit circulating levels of free haemoglobin, haem and iron - potentially toxic species that are released from injured RBCs. However, these adaptive defence systems can fail owing to ongoing intravascular disintegration of RBCs. Induction of the haem-degrading enzyme haem oxygenase 1 (HO1) - and potentially HO2 - represents a response to, and endogenous defence against, large amounts of cellular haem; however, this system can also become saturated. A frequent adverse consequence of massive and/or chronic haemolysis is kidney injury, which contributes to the morbidity and mortality of chronic haemolytic diseases. Intravascular destruction of RBCs and the resulting accumulation of haemoproteins can induce kidney injury via a number of mechanisms, including oxidative stress and cytotoxicity pathways, through the formation of intratubular casts and through direct as well as indirect proinflammatory effects, the latter via the activation of neutrophils and monocytes. Understanding of the detailed pathophysiology of haemolysis-induced kidney injury offers opportunities for the design and implementation of new therapeutic strategies to counteract the unfavourable and potentially fatal effects of haemolysis on the kidney.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany.
| | - Erfan Nur
- Department of Haematology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Department of Haematology and Central Haematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
12
|
Tóth A, Berente Z, Bogner P, Környei B, Balogh B, Czeiter E, Amrein K, Dóczi T, Büki A, Schwarcz A. Cerebral Microbleeds Temporarily Become Less Visible or Invisible in Acute Susceptibility Weighted Magnetic Resonance Imaging: A Rat Study. J Neurotrauma 2019; 36:1670-1677. [PMID: 30421664 PMCID: PMC6531906 DOI: 10.1089/neu.2018.6004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Previously, we reported human traumatic brain injury cases demonstrating acute to subacute microbleed appearance changes in susceptibility-weighted imaging (SWI—magnetic resonance imaging [MRI]). This study aims to confirm and characterize such temporal microbleed appearance alterations in an experimental model. To elicit microbleed formation, brains of male Sprague Dawley rats were pierced in a depth of 4 mm, in a parasagittal position bilaterally using 159 μm and 474 μm needles, without the injection of autologous blood or any agent. Rats underwent 4.7 T MRI immediately, then at multiple time points until 125 h. Volumes of hypointensities consistent with microbleeds in SWI were measured using an intensity threshold-based approach. Microbleed volumes across time points were compared using repeated measures analysis of variance. Microbleeds were assessed by Prussian blue histology at different time points. Hypointensity volumes referring to microbleeds were significantly decreased (corrected p < 0.05) at 24 h compared with the immediate or the 125 h time points. By visual inspection, microbleeds were similarly detectable at the immediate and 125 h imaging but were decreased in extent or completely absent at 24 h or 48 h. Histology confirmed the presence of microbleeds at all time points and in all animals. This study confirmed a general temporary reduction in visibility of microbleeds in the acute phase in SWI. Such short-term appearance dynamics of microbleeds should be considered when using SWI as a diagnostic tool for microbleeds in traumatic brain injury and various diseases.
Collapse
Affiliation(s)
- Arnold Tóth
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary.,2 Department of Radiology, Pécs Medical School, Pécs, Hungary.,3 MTA-PTE Clinical Neuroscience MR Research Group, Pécs, Hungary
| | - Zoltán Berente
- 4 Department of Biochemistry and Medical Chemistry, Pécs Medical School, Pécs, Hungary.,5 János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,6 Research Group for Experimental Diagnostic Imaging, Pécs Medical School, Pécs, Hungary
| | - Péter Bogner
- 2 Department of Radiology, Pécs Medical School, Pécs, Hungary
| | - Bálint Környei
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary
| | - Bendegúz Balogh
- 2 Department of Radiology, Pécs Medical School, Pécs, Hungary
| | - Endre Czeiter
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary.,3 MTA-PTE Clinical Neuroscience MR Research Group, Pécs, Hungary.,5 János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Krisztina Amrein
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary.,5 János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Tamás Dóczi
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary.,3 MTA-PTE Clinical Neuroscience MR Research Group, Pécs, Hungary.,7 Diagnostic Center of Pécs, Pécs, Hungary
| | - András Büki
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary.,5 János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Attila Schwarcz
- 1 Department of Neurosurgery, Pécs Medical School, Pécs, Hungary
| |
Collapse
|
13
|
Tyrosine residues of bovine serum albumin play an important role in protecting SH-SY5Y cells against heme/H2O2/NO2−-induced damage. Mol Cell Biochem 2018; 454:57-66. [DOI: 10.1007/s11010-018-3452-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/05/2018] [Indexed: 02/08/2023]
|
14
|
Huang LC, Liew HK, Cheng HY, Kuo JS, Hsu WL, Pang CY. Brain Magnetic Resonance Imaging of Intracerebral Hemorrhagic Rats after Alcohol Consumption. J Stroke Cerebrovasc Dis 2018; 27:3493-3502. [PMID: 30205999 DOI: 10.1016/j.jstrokecerebrovasdis.2018.08.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/23/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Alcoholism is one of the risk factors for cerebrovascular diseases. Our previous study demonstrated that acute alcohol intoxication enhances brain injury and neurological impairment in rats suffering from intracerebral hemorrhage (ICH). We plan to investigate the effect of chronic alcohol consumption (CAC) in rats with ICH by magnetic resonance imaging (MRI). METHODS Sixteen Sprague-Dawley male rats were divided into 2 groups: CAC group (fed with 10% alcohol drinking water for 4 weeks, n = 8), and Control group (plain drinking water, n = 8). ICH was induced by collagenase infusion into the right striata of all rats. Coronal T1-weighted imaging, T2-weighted imaging, T2*-weighted imaging, and diffusion-weighted imaging were generated with a 3.0T MRI scanner to investigate the changes of hemorrhagic volume and edema throughout the injury and recovery stages of ICH in rats. RESULTS T2-weighted imaging is ideal for monitoring hematoma volume in rats. The hematoma volume was larger in the CAC group than in the control group (P < .001), however, did not correlate to post-ICH progressive edema formation (P > .7), and neurological impairment (P > .28) between the 2 groups, respectively. DISCUSSION Although our findings indicate that CAC induces larger hematoma in rats with ICH, the underlying mechanism should be studied in the future.
Collapse
Affiliation(s)
- Li-Chuan Huang
- Department of Radiology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Hock-Kean Liew
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Cardiovascular and Metabolomics Research Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Hung-Yu Cheng
- Department of Physical Medicine and Rehabilitation, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Jon-Son Kuo
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Master Program and PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Wen-Lin Hsu
- Department of Radiation Oncology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Cheng-Yoong Pang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan; Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan; Cardiovascular and Metabolomics Research Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.
| |
Collapse
|
15
|
Cao S, Hua Y, Keep RF, Chaudhary N, Xi G. Minocycline Effects on Intracerebral Hemorrhage-Induced Iron Overload in Aged Rats: Brain Iron Quantification With Magnetic Resonance Imaging. Stroke 2018; 49:995-1002. [PMID: 29511126 DOI: 10.1161/strokeaha.117.019860] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/05/2018] [Accepted: 01/25/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND PURPOSE Brain iron overload is a key factor causing brain injury after intracerebral hemorrhage (ICH). This study quantified brain iron levels after ICH with magnetic resonance imaging R2* mapping. The effect of minocycline on iron overload and ICH-induced brain injury in aged rats was also determined. METHODS Aged (18 months old) male Fischer 344 rats had an intracerebral injection of autologous blood or saline, and brain iron levels were measured by magnetic resonance imaging R2* mapping. Some ICH rats were treated with minocycline or vehicle. The rats were euthanized at days 7 and 28 after ICH, and brains were used for immunohistochemistry and Western blot analyses. Magnetic resonance imaging (T2-weighted, T2* gradient-echo, and R2* mapping) sequences were performed at different time points. RESULTS ICH-induced brain iron overload in the perihematomal area could be quantified by R2* mapping. Minocycline treatment reduced brain iron accumulation, T2* lesion volume, iron-handling protein upregulation, neuronal cell death, and neurological deficits (P<0.05). CONCLUSIONS Magnetic resonance imaging R2* mapping is a reliable and noninvasive method, which can quantitatively measure brain iron levels after ICH. Minocycline reduced ICH-related perihematomal iron accumulation and brain injury in aged rats.
Collapse
Affiliation(s)
- Shenglong Cao
- From the Department of Neurosurgery (S.C., Y.H., R.F.K., N.C., G.X.) and Department of Radiology (N.C.), University of Michigan, Ann Arbor; and Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C.)
| | - Ya Hua
- From the Department of Neurosurgery (S.C., Y.H., R.F.K., N.C., G.X.) and Department of Radiology (N.C.), University of Michigan, Ann Arbor; and Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C.)
| | - Richard F Keep
- From the Department of Neurosurgery (S.C., Y.H., R.F.K., N.C., G.X.) and Department of Radiology (N.C.), University of Michigan, Ann Arbor; and Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C.)
| | - Neeraj Chaudhary
- From the Department of Neurosurgery (S.C., Y.H., R.F.K., N.C., G.X.) and Department of Radiology (N.C.), University of Michigan, Ann Arbor; and Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C.)
| | - Guohua Xi
- From the Department of Neurosurgery (S.C., Y.H., R.F.K., N.C., G.X.) and Department of Radiology (N.C.), University of Michigan, Ann Arbor; and Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China (S.C.).
| |
Collapse
|
16
|
Di Napoli M, Behrouz R, Topel CH, Misra V, Pomero F, Giraudo A, Pennati P, Masotti L, Schreuder FHBM, Staals J, Klijn CJM, Smith CJ, Parry-Jones AR, Slevin MA, Silver B, Willey JZ, Azarpazhooh MR, Vallejo JM, Nzwalo H, Popa-Wagner A, Godoy DA. Hypoalbuminemia, systemic inflammatory response syndrome, and functional outcome in intracerebral hemorrhage. J Crit Care 2017; 41:247-253. [PMID: 28599198 DOI: 10.1016/j.jcrc.2017.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/23/2017] [Accepted: 06/01/2017] [Indexed: 01/25/2023]
Abstract
PURPOSE Hypoalbuminemia and systemic inflammatory response syndrome (SIRS) are reported in critically-ill patients, but their relationship is unclear. We sought to determine the association of admission serum albumin and SIRS with outcomes in patients with intracerebral hemorrhage (ICH). METHODS We used a multicenter, multinational registry of ICH patients to select patients in whom SIRS parameters and serum albumin levels had been determined on admission. Hypoalbuminemia was defined as the lowest standardized quartile of albumin; SIRS according to standard criteria. Primary outcomes were modified Rankin Scale (mRS) at discharge and in-hospital mortality. Regression models were used to assess for the association of hypoalbuminemia and SIRS with discharge mRS and in-hospital mortality. RESULTS Of 761 ICH patients included in the registry 518 met inclusion criteria; 129 (25%) met SIRS criteria on admission. Hypoalbuminemia was more frequent in patients with SIRS (42% versus 19%; p<0.001). SIRS was associated with worse outcomes (OR: 4.68, 95%CI, 2.52-8.76) and in-hospital all-cause mortality (OR: 2.18, 95% CI, 1.60-2.97), while hypoalbuminemia was not associated with all-cause mortality. CONCLUSIONS In patients with ICH, hypoalbuminemia is strongly associated with SIRS. SIRS, but not hypoalbuminemia, predicts poor outcome at discharge. Recognizing and managing SIRS early may prevent death or disability in ICH patients.
Collapse
Affiliation(s)
- Mario Di Napoli
- Neurological Service, San Camillo de' Lellis General Hospital, Rieti, Italy; Neurological Section, Neuro-epidemiology Unit, SMDN, Centre for Cardiovascular Medicine and Cerebrovascular Disease Prevention, Sulmona, L'Aquila, Italy.
| | - Réza Behrouz
- Department of Neurology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Christopher H Topel
- Department of Neurology, School of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Vivek Misra
- Houston Methodist Hospital, Houston, TX, USA
| | - Fulvio Pomero
- Department of Internal Medicine, Ospedale Santa Croce e Carle, Cuneo, Italy
| | - Alessia Giraudo
- Department of Internal Medicine, Ospedale Santa Croce e Carle, Cuneo, Italy
| | - Paolo Pennati
- Department of Internal Medicine, Ospedale Santa Croce e Carle, Cuneo, Italy; Emergency-Urgency Department, Ospedale Livorno, Livorno, Italy
| | - Luca Masotti
- Department of Internal Medicine, Ospedale Santa Maria Nuova, Florence, Italy
| | - Floris H B M Schreuder
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Catharina J M Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Craig J Smith
- Stroke and Vascular Centre, Institute of Cardiovascular Sciences, University of Manchester, UK; Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Stott Lane, Salford, UK
| | - Adrian R Parry-Jones
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Mark A Slevin
- School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Brian Silver
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Joshua Z Willey
- Department of Neurology, Columbia University College of Physicians & Surgeons, New York, NY, USA
| | - Mahmoud R Azarpazhooh
- Department of Neurology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | | - Hipólito Nzwalo
- Stroke Unit, Centro Hospitalar do Algarve, University do Algarve, Algarve, Portugal
| | - Aurel Popa-Wagner
- University of Medicine and Pharmacy Craiova, Romania; Department of Psychiatry, Universitätsmedizin Rostock, Rostock, Germany
| | - Daniel A Godoy
- Neurocritical Care Unit, Sanatorio Pasteur, Catamarca, Argentina; Unidad de Terapia Intensiva, Hospital Interzonal de Agudos "San Juan Bautista", Catamarca, Argentina
| | | |
Collapse
|
17
|
Obenaus A, Ng M, Orantes AM, Kinney-Lang E, Rashid F, Hamer M, DeFazio RA, Tang J, Zhang JH, Pearce WJ. Traumatic brain injury results in acute rarefication of the vascular network. Sci Rep 2017; 7:239. [PMID: 28331228 PMCID: PMC5427893 DOI: 10.1038/s41598-017-00161-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 02/13/2017] [Indexed: 01/04/2023] Open
Abstract
The role of the cerebrovascular network and its acute response to TBI is poorly defined and emerging evidence suggests that cerebrovascular reactivity is altered. We explored how cortical vessels are physically altered following TBI using a newly developed technique, vessel painting. We tested our hypothesis that a focal moderate TBI results in global decrements to structural aspects of the vasculature. Rats (naïve, sham-operated, TBI) underwent a moderate controlled cortical impact. Animals underwent vessel painting perfusion to label the entire cortex at 1 day post TBI followed by whole brain axial and coronal images using a wide-field fluorescence microscope. Cortical vessel network characteristics were analyzed for classical angiographic features (junctions, lengths) wherein we observed significant global (both hemispheres) reductions in vessel junctions and vessel lengths of 33% and 22%, respectively. Biological complexity can be quantified using fractal geometric features where we observed that fractal measures were also reduced significantly by 33%, 16% and 13% for kurtosis, peak value frequency and skewness, respectively. Acutely after TBI there is a reduction in vascular network and vascular complexity that are exacerbated at the lesion site and provide structural evidence for the bilateral hemodynamic alterations that have been reported in patients after TBI.
Collapse
Affiliation(s)
- Andre Obenaus
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| | - Michelle Ng
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Amanda M Orantes
- Molecular and Integrative Physiology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Eli Kinney-Lang
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Faisal Rashid
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Mary Hamer
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | | | - Jiping Tang
- Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - William J Pearce
- Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, 92350, USA
| |
Collapse
|
18
|
De Reuck J, Cordonnier C, Deramecourt V, Auger F, Durieux N, Leys D, Pasquier F, Maurage CA, Bordet R. Lobar intracerebral haematomas: Neuropathological and 7.0-tesla magnetic resonance imaging evaluation. J Neurol Sci 2016; 369:121-125. [PMID: 27653876 DOI: 10.1016/j.jns.2016.08.012] [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: 07/02/2016] [Revised: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND AND PURPOSE The Boston criteria for cerebral amyloid angiopathy (CAA) need validation by neuropathological examination in patients with lobar cerebral haematomas (LCHs). In "vivo" 1.5-tesla magnetic resonance imaging (MRI) is unreliable to detect the age-related signal changes in LCHs. This post-mortem study investigates the validity of the Boston criteria in brains with LCHs and the signal changes during their time course with 7.0-tesla MRI. MATERIALS AND METHODS Seventeen CAA brains including 26 LCHs were compared to 13 non-CAA brains with 14 LCHs. The evolution of the signal changes with time was examined in 25 LCHs with T2 and T2* 7.0-tesla MRI. RESULTS In the CAA group LCHs were predominantly located in the parieto-occipital lobes. Also white matter changes were more severe with more cortical microinfarcts and cortical microbleeds. On MRI there was a progressive shift of the intensity of the hyposignal from the haematoma core in the acute stage to the boundaries later on. During the residual stage the hyposignal mildly decreased in the boundaries with an increase of the superficial siderosis and haematoma core collapse. CONCLUSIONS Our post-mortem study of LCHs confirms the validity of the Boston criteria for CAA. Also 7.0-tesla MRI allows staging the age of the LCHs.
Collapse
Affiliation(s)
- Jacques De Reuck
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France.
| | - Charlotte Cordonnier
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Vincent Deramecourt
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Florent Auger
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Nicolas Durieux
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Didier Leys
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Florence Pasquier
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Claude-Alain Maurage
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| | - Regis Bordet
- Université Lille 2, INSERM U1171, Degenerative & vascular cognitive disorders, CHU Lille, F-59000 Li, France
| |
Collapse
|
19
|
Pearce WJ, Doan C, Carreon D, Kim D, Durrant LM, Manaenko A, McCoy L, Obenaus A, Zhang JH, Tang J. Imatinib attenuates cerebrovascular injury and phenotypic transformation after intracerebral hemorrhage in rats. Am J Physiol Regul Integr Comp Physiol 2016; 311:R1093-R1104. [PMID: 27707720 DOI: 10.1152/ajpregu.00240.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/15/2016] [Accepted: 09/25/2016] [Indexed: 12/27/2022]
Abstract
This study explored the hypothesis that intracerebral hemorrhage (ICH) promotes release of diffusible factors that can significantly influence the structure and function of cerebral arteries remote from the site of injury, through action on platelet-derived growth factor (PDGF) receptors. Four groups of adult male Sprague-Dawley rats were studied (n = 8 each): 1) sham; 2) sham + 60 mg/kg ip imatinib; 3) ICH (collagenase method); and 4) ICH + 60 mg/kg ip imatinib given 60 min after injury. At 24 h after injury, sham artery passive diameters (+3 mM EGTA) averaged 244 ± 7 µm (at 60 mmHg). ICH significantly increased passive diameters up to 6.4% and decreased compliance up to 42.5%. For both pressure- and potassium-induced contractions, ICH decreased calcium mobilization up to 26.2% and increased myofilament calcium sensitivity up to 48.4%. ICH reduced confocal colocalization of smooth muscle α-actin (αActin) with nonmuscle myosin heavy chain (MHC) and increased its colocalization with smooth muscle MHC, suggesting that ICH promoted contractile differentiation. ICH also enhanced colocalization of myosin light chain kinase (MLCK) with both αActin and regulatory 20-kDa myosin light chain. All effects of ICH on passive diameter, compliance, contractility, and contractile protein colocalization were significantly reduced or absent in arteries from animals treated with imatinib. These findings support the hypothesis that ICH promotes release into the cerebrospinal fluid of vasoactive factors that can diffuse to and promote activation of cerebrovascular PDGF receptors, thereby altering the structure, contractile protein organization, contractility, and smooth muscle phenotype of cerebral arteries remote from the site of hemorrhage.
Collapse
Affiliation(s)
- William J Pearce
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California; .,Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Coleen Doan
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California.,Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Desirelys Carreon
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California.,Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Dahlim Kim
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California.,Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Lara M Durrant
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California.,Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California
| | - Anatol Manaenko
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| | - Lauren McCoy
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California; and
| | - Andre Obenaus
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California; and
| | - John H Zhang
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, California.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Department of Pharmacology, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
20
|
Tao C, Zhang R, Hu X, Song L, Wang C, Gao F, You C. A Novel Brainstem Hemorrhage Model by Autologous Blood Infusion in Rat: White Matter Injury, Magnetic Resonance Imaging, and Neurobehavioral Features. J Stroke Cerebrovasc Dis 2016; 25:1102-1109. [DOI: 10.1016/j.jstrokecerebrovasdis.2016.01.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/20/2016] [Indexed: 12/16/2022] Open
|
21
|
Toth A, Kovacs N, Tamas V, Kornyei B, Nagy M, Horvath A, Rostas T, Bogner P, Janszky J, Doczi T, Buki A, Schwarcz A. Microbleeds may expand acutely after traumatic brain injury. Neurosci Lett 2016; 617:207-12. [PMID: 26912192 DOI: 10.1016/j.neulet.2016.02.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/14/2016] [Accepted: 02/15/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND PURPOSE Susceptibility weighted imaging (SWI) is a very sensitive tool for the detection of microbleeds in traumatic brain injury (TBI). The number and extent of such traumatic microbleeds (TMBs) have been shown to correlate with the severity of the injury and the clinical outcome. However, the acute dynamics of TMBs have not been revealed so far. Since TBI is known to constitute dynamic pathological processes, we hypothesized that TMBs are not constant in their appearance, but may progress acutely after injury. MATERIALS AND METHODS We present here five closed moderate/severe (Glasgow coma scale≤13) TBI patients who underwent SWI very early (average=23.4 h), and once again a week (average=185.8 h) after the injury. The TMBs were mapped at both time points by a conventional radiological approach and their numbers and volumes were measured with manual tracing tools by two observers. TMB counts and extents were compared between time points. RESULTS TMBs were detected in four patients, three of them displaying an apparent TMB change. In these patients, TMB confluence and apparent growth were detected in the corpus callosum, coronal radiation or subcortical white matter, while unchanged TMBs were also present. These changes caused a decrease in the TMB count associated with an increase in the overall TMB volume over time. CONCLUSION We have found a compelling evidence that diffuse axonal injury-related microbleed development is not limited strictly to the moment of injury: the TMBs might expand in the acute phase of TBI. The timing of SWI acquisition may be relevant for optimizing the prognostic utility of this imaging biomarker.
Collapse
Affiliation(s)
- Arnold Toth
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary.
| | - Noemi Kovacs
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary.
| | - Viktoria Tamas
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary.
| | - Balint Kornyei
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary.
| | - Mate Nagy
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary.
| | - Andrea Horvath
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary; Diagnostic Center of Pécs, H-7623, Rét. u. 2., Pécs, Hungary.
| | - Tamas Rostas
- Department of Radiology, Pécs Medical School, H-7624, Ifjusag str. 13., Pécs, Hungary.
| | - Peter Bogner
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary; Department of Radiology, Pécs Medical School, H-7624, Ifjusag str. 13., Pécs, Hungary.
| | - Jozsef Janszky
- Department of Neurology, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary; MTA-PTE Clinical Neuroscience MR Research Group, Hungary.
| | - Tamas Doczi
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary; Diagnostic Center of Pécs, H-7623, Rét. u. 2., Pécs, Hungary; MTA-PTE Clinical Neuroscience MR Research Group, Hungary.
| | - Andras Buki
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary; MTA-PTE Clinical Neuroscience MR Research Group, Hungary.
| | - Attila Schwarcz
- Department of Neurosurgery, Pécs Medical School, H-7623, Rét. u. 2., Pécs, Hungary; MTA-PTE Clinical Neuroscience MR Research Group, Hungary.
| |
Collapse
|
22
|
Xu Z, Li X, Chen J, Zhao J, Wang J, Ji Y, Shen Y, Han L, Shi J, Zhang D. USP11, Deubiquitinating Enzyme, Associated with Neuronal Apoptosis Following Intracerebral Hemorrhage. J Mol Neurosci 2015; 58:16-27. [PMID: 26334325 DOI: 10.1007/s12031-015-0644-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 08/18/2015] [Indexed: 11/29/2022]
Abstract
Protein ubiquitination is a dynamic two-way process that can be reversed or regulated by deubiquitinating enzymes (DUB). USP11, located on the X chromosome, 6 is a member of USP subclass of the DUB family. Here, we demonstrate that USP11 may be involved in neuronal apoptosis in the processes of intracerebral hemorrhage (ICH). From the results of Western blot, immunohistochemistry, and immunofluorescence, we obtained a significant up-regulation of USP11 in neurons adjacent to the hematoma following ICH. Increasing USP11 level was found to be accompanied by the up-regulation of active caspase-3, Fas receptor (Fas), Fas ligand (FasL), and active caspase-8. Besides, USP11 co-localized well with active caspase-3 in neurons, indicating its potential role in neuronal apoptosis. What is more, knocking down USP11 by RNA-interference in PC12 cells reduced active caspase-3 expression. Thus, USP11 may play a role in promoting the brain secondary damage following ICH.
Collapse
Affiliation(s)
- Zhiwei Xu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Xiaohong Li
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jianping Chen
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jianmei Zhao
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Jun Wang
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Yuhong Ji
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Yifen Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Lijian Han
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Jiansheng Shi
- Surgical Comprehensive Laboratory, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China.
| | - Dongmei Zhang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
23
|
Toxic role of prostaglandin E2 receptor EP1 after intracerebral hemorrhage in mice. Brain Behav Immun 2015; 46:293-310. [PMID: 25697396 PMCID: PMC4422065 DOI: 10.1016/j.bbi.2015.02.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 01/28/2015] [Accepted: 02/08/2015] [Indexed: 02/07/2023] Open
Abstract
Inflammatory mechanisms mediated by prostaglandins may contribute to the progression of intracerebral hemorrhage (ICH)-induced brain injury, but they are not fully understood. In this study, we examined the effect of prostaglandin E2 receptor EP1 (EP1R) activation and inhibition on brain injury in mouse models of ICH and investigated the underlying mechanism of action. ICH was induced by injecting collagenase, autologous blood, or thrombin into the striatum of middle-aged male and female mice and aged male mice. Effects of selective EP1R agonist ONO-DI-004, antagonist SC51089, and nonspecific Src family kinase inhibitor PP2 were evaluated by a combination of histologic, magnetic resonance imaging (MRI), immunofluorescence, molecular, cellular, and behavioral assessments. EP1R was expressed primarily in neurons and axons but not in astrocytes or microglia after ICH induced by collagenase. In middle-aged male mice subjected to collagenase-induced ICH, EP1R inhibition mitigated brain injury, brain edema, cell death, neuronal degeneration, neuroinflammation, and neurobehavioral deficits, whereas its activation exacerbated these outcomes. EP1R inhibition also was protective in middle-aged female mice and aged male mice after collagenase-induced ICH and in middle-aged male mice after blood- or thrombin-induced ICH. EP1R inhibition also reduced oxidative stress, white matter injury, and brain atrophy and improved functional outcomes. Histologic results were confirmed by MRI. Src kinase phosphorylation and matrix metalloproteinase-9 activity were increased by EP1R activation and decreased by EP1R inhibition. EP1R regulated matrix metalloproteinase-9 activity through Src kinase signaling, which mediated EP1R toxicity after collagenase-induced ICH. We conclude that prostaglandin E2 EP1R activation plays a toxic role after ICH through mechanisms that involve the Src kinases and the matrix metalloproteinase-9 signaling pathway. EP1R inhibition could be a novel therapeutic strategy to improve outcomes after ICH.
Collapse
|
24
|
Egashira Y, Hua Y, Keep RF, Xi G. Intercellular cross-talk in intracerebral hemorrhage. Brain Res 2015; 1623:97-109. [PMID: 25863131 DOI: 10.1016/j.brainres.2015.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 12/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating cerebrovascular disorder with high mortality and morbidity. Currently, there are few treatment strategies for ICH-induced brain injury. A recent increase in interest in the pathophysiology of ICH has led to elucidation of the pathways underlying ICH-induced brain injury, pathways where intercellular and hematoma to cell signaling play important roles. In this review, we summarize recent advances in ICH research focusing on intercellular and hematoma:cell cross-talk related to brain injury and recovery after ICH. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yusuke Egashira
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Sonni S, Lioutas VA, Selim MH. New avenues for treatment of intracranial hemorrhage. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2013; 16:277. [PMID: 24366522 DOI: 10.1007/s11936-013-0277-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OPINION STATEMENT The mortality and morbidity from intracerebral hemorrhage (ICH) remain high despite advances in medical, neurologic, and surgical care during the past decade. The lessons learned from previous therapeutic trials in ICH, improved understanding of the pathophysiology of neuronal injury after ICH, and advances in imaging and pre-hospital assessment technologies provide optimism that more effective therapies for ICH are likely to emerge in the coming years. The potential new avenues for the treatment of ICH include a combination of increased utilization of minimally invasive surgical techniques with or without thrombolytic usage to evacuate or reduce the size of the hematoma; utilization of advanced imaging to improve selection of patients who are likely to benefit from reversal of coagulopathy or hemostatic therapy; ultra-early diagnosis and initiation of therapy in the ambulance; and the use of novel drugs to target the secondary injury mechanisms, including the inflammatory cascade, perihematomal edema reduction, and hemoglobin degradation products-mediated toxicity.
Collapse
Affiliation(s)
- Shruti Sonni
- Department of Neurology, Cambridge Hospital, 1493 Cambridge Street, Cambridge, MA, 02139, USA,
| | | | | |
Collapse
|
26
|
Ewen T, Qiuting L, Chaogang T, Tao T, Jun W, Liming T, Guanghong X. Neuroprotective effect of atorvastatin involves suppression of TNF-α and upregulation of IL-10 in a rat model of intracerebral hemorrhage. Cell Biochem Biophys 2013; 66:337-46. [PMID: 23090788 DOI: 10.1007/s12013-012-9453-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We evaluated the neuroprotective effects of atorvastatin (2, 5, and 10 mg/kg) on experimentally induced intracerebral hemorrhage (ICH) in adult rats; controls were administered PBS. Plasma TNF-α and IL-10 levels before and after ICH were analyzed at various time points by enzyme-linked immunosorbent assay (ELISA) and neurological behavior of rats was assessed by climbing scores. At 3-days postoperatively, brain water contents and TNF-α/IL-10 expression in brain tissue were determined. Histopathological changes and microglial cells in the brain tissue were evaluated by light-microscopy. Post-ICH neurological deficits differed significantly between sham-operated group A and experimental-ICH group B (P < 0.05). Brain water contents were significantly less in group A than in group B (P < 0.05). Significant differences (P < 0.05) between two groups were observed regarding activated microglia, TNF-α and IL-10 levels. Compared with group B, neurological deficits, brain water contents, pathological changes, and activated microglia were reduced (P < 0.05) in groups C (Experimental-ICH + atorvastatin 2 mg/kg), D (Experimental-ICH + atorvastatin 5 mg/kg) and E (Experimental-ICH + atorvastatin 10 mg/kg). Atorvastatin-induced a dose-dependent reduction of TNF-α and increase of IL-10 levels (P < 0.05). Therefore, it was concluded that atorvastatin improved neurofunctional rehabilitation in rats through the suppression of cytokines-mediated inflammatory response and attenuation of brain damage following intracerebral hemorrhage.
Collapse
Affiliation(s)
- Tu Ewen
- Department of Neurology, The Brains Hospital of Hunan Province, Changsha, 410007, Hunan Province, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Turtzo LC, Budde MD, Gold EM, Lewis BK, Janes L, Yarnell A, Grunberg NE, Watson W, Frank JA. The evolution of traumatic brain injury in a rat focal contusion model. NMR IN BIOMEDICINE 2013; 26:468-479. [PMID: 23225324 PMCID: PMC3596464 DOI: 10.1002/nbm.2886] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 08/28/2012] [Accepted: 10/05/2012] [Indexed: 06/01/2023]
Abstract
Serial MRI facilitates the in vivo analysis of the intra- and intersubject evolution of traumatic brain injury lesions. Despite the availability of MRI, the natural history of experimental focal contusion lesions in the controlled cortical impact (CCI) rat model has not been well described. We performed CCI on rats and MRI during the acute to chronic stages of cerebral injury to investigate the time course of changes in the brain. Female Wistar rats underwent CCI of their left motor cortex with a flat impact tip driven by an electromagnetic piston. In vivo MRI was performed at 7 T serially over 6 weeks post-CCI. The appearances of CCI-induced lesions and lesion-associated cortical volumes were variable on MRI, with the percentage change in cortical volume of the CCI ipsilateral side relative to the contralateral side ranging from 18% within 2 h of injury on day 0 to a peak of 35% on day 1, and a trough of -28% by week 5/6, with an average standard deviation of ± 14% at any given time point. In contrast, the percentage change in cortical volume of the ipsilateral side relative to the contralateral side in control rats was not significant (1 ± 2%). Hemorrhagic conversion within and surrounding the CCI lesion occurred between days 2 and 9 in 45% of rats, with no hemorrhage noted on the initial scan. Furthermore, hemorrhage and hemosiderin within the lesion were positive for Prussian blue and highly autofluorescent on histological examination. Although some variation in injuries may be technique related, the divergence of similar lesions between initial and final scans demonstrates the inherent biological variability of the CCI rat model.
Collapse
Affiliation(s)
- L. Christine Turtzo
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D. Budde
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Eric M. Gold
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Bobbi K. Lewis
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Janes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Angela Yarnell
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Neil E. Grunberg
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - William Watson
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joseph A. Frank
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
28
|
Jin H, Wu G, Hu S, Hua Y, Keep RF, Wu J, Xi G. T2 and T2* magnetic resonance imaging sequences predict brain injury after intracerebral hemorrhage in rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 118:151-5. [PMID: 23564123 DOI: 10.1007/978-3-7091-1434-6_28] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Magnetic resonance imaging (MRI) has been widely used in intracerebral hemorrhage (ICH) animal models and patients. In the current study, we examined whether MRI can predict at-risk brain tissue during the acute phase and long-term brain tissue loss after ICH. Male Sprague-Dawley rats had an intracaudate injection of autologous whole blood (10, 50 or 100 μL). MRI (T2 and T2*) sequences were performed at days 1, 3, 7, 14, and 28. The volume of brain tissue at risk was calculated as the difference between T2 and T2* lesion volumes. Dopamine- and cAMP-regulated phosphoprotein, Mr 32 kDa (DARPP-32) was used as a neuronal marker in the basal ganglia. Brain swelling at day 3 and brain tissue loss at day 28 after ICH were also measured. We found that the difference in lesion volumes between T2 and T2* measured by MRI coincided well with the difference between the volume of the DARPP-32-negative area and that of the hematoma measured in brain sections. Volumes of brain tissue at risk at day 3 correlated with the brain swelling at day 3 (p < 0.01) as well as the final brain tissue loss at day 28 (n = 9, p < 0.05). The results suggest that the difference between T2 lesions and T2* lesions could be an indicator of at-risk brain tissue and it could be used as a predictor of neuronal loss in ICH patients.
Collapse
Affiliation(s)
- Hang Jin
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Babu R, Bagley JH, Di C, Friedman AH, Adamson C. Thrombin and hemin as central factors in the mechanisms of intracerebral hemorrhage-induced secondary brain injury and as potential targets for intervention. Neurosurg Focus 2012; 32:E8. [PMID: 22463118 DOI: 10.3171/2012.1.focus11366] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stoke that may cause significant morbidity and mortality. Brain injury due to ICH initially occurs within the first few hours as a result of mass effect due to hematoma formation. However, there is increasing interest in the mechanisms of secondary brain injury as many patients continue to deteriorate clinically despite no signs of rehemorrhage or hematoma expansion. This continued insult after primary hemorrhage is believed to be mediated by the cytotoxic, excitotoxic, oxidative, and inflammatory effects of intraparenchymal blood. The main factors responsible for this injury are thrombin and erythrocyte contents such as hemoglobin. Therapies including thrombin inhibitors, N-methyl-D-aspartate antagonists, chelators to bind free iron, and antiinflammatory drugs are currently under investigation for reducing this secondary brain injury. This review will discuss the molecular mechanisms of brain injury as a result of intraparenchymal blood, potential targets for therapeutic intervention, and treatment strategies currently in development.
Collapse
Affiliation(s)
- Ranjith Babu
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | |
Collapse
|
30
|
Eady TN, Khoutorova L, Atkins KD, Bazan NG, Belayev L. Docosahexaenoic acid complexed to human albumin in experimental stroke: neuroprotective efficacy with a wide therapeutic window. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:19. [PMID: 22980673 PMCID: PMC3540001 DOI: 10.1186/2040-7378-4-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 09/06/2012] [Indexed: 06/01/2023]
Abstract
BACKGROUND Docosahexaenoic acid (DHA) complexed to human serum albumin (Alb) is neuroprotective after experimental stroke. Here we tested using lower concentrations of albumin as part of the complex to achieve neuroprotection. We found that lower Alb concentrations extend the therapeutic window of protection beyond 5 h after stroke onset. METHODS Sprague-Dawley rats were received 2 h middle cerebral artery occlusion (MCAo). The behavior was evaluated on day 1, 2, 3 and 7 after MCAo. In the dose-response study, animals were given either DHA (5mg/kg), Alb (0.63g/kg), DHA-Alb (5mg/kg + 0.32, 0.63 or 1.25 g/kg) or saline, i.v. 3 h after onset of stroke (n=6-8 per group). In the therapeutic window study, DHA-Alb (5mg/kg + 1.25g/kg) was administered i.v. at either 3, 4, 5, 6 or 7 h after onset of stroke (n=7-9 per group). Alb (1.25g/kg) was given at 3 h or 5 h and saline at 3h after onset of reperfusion. Seven days after MCAo, infarct volumes and number of GFAP, ED-1, NeuN, SMI-71 positive cells and vessels were counted. RESULTS Moderate DHA-Alb doses (0.63 and 1.25 g/kg) improved neurological scores compared to albumin-treated rats on days 1, 2, 3 and 7. All DHA-Alb doses (0.32, 0.63 and 1.25 g/kg) markedly reduced cortical (by 65-70%), striatal (by 52-63%) and total infarct volumes (by 60-64%) compared to native Alb group. In the therapeutic window study DHA-Alb led to improved neurological score and significant reductions of infarct volumes (especially in the cortical or penumbral region), even when treatment was initiated as late as 7 hours after onset of MCAo. CONCLUSIONS The DHA-Alb complex affords high-grade neurobehavioral neuroprotection in focal cerebral ischemia, equaling or exceeding that afforded by native Alb or DHA, at considerably moderate doses. It has a broad therapeutic window extending to 7 h after stroke onset. Taken together, these finding support the potential clinical feasibility of administering DHA-Alb therapy to patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Tiffany N Eady
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Kristal D Atkins
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Ludmila Belayev
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
- Department of Neurosurgery, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New 2020 Gravier Street, Suite D, Orleans, LA, 70112, USA
| |
Collapse
|
31
|
Computational analysis reveals increased blood deposition following repeated mild traumatic brain injury. NEUROIMAGE-CLINICAL 2012; 1:18-28. [PMID: 24179733 PMCID: PMC3757717 DOI: 10.1016/j.nicl.2012.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/12/2012] [Accepted: 08/04/2012] [Indexed: 11/22/2022]
Abstract
Mild traumatic brain injury (mTBI) has become an increasing public health concern as subsequent injuries can exacerbate existing neuropathology and result in neurological deficits. This study investigated the temporal development of cortical lesions using magnetic resonance imaging (MRI) to assess two mTBIs delivered to opposite cortical hemispheres. The controlled cortical impact model was used to produce an initial mTBI on the right cortex followed by a second injury induced on the left cortex at 3 (rmTBI 3d) or 7 (rmTBI 7d) days later. Histogram analysis was combined with a novel semi-automated computational approach to perform a voxel-wise examination of extravascular blood and edema volumes within the lesion. Examination of lesion volume 1d post last injury revealed increased tissue abnormalities within rmTBI 7d animals compared to other groups, particularly at the site of the second impact. Histogram analysis of lesion T2 values suggested increased edematous tissue within the rmTBI 3d group and elevated blood deposition in the rm TBI 7d animals. Further quantification of lesion composition for blood and edema containing voxels supported our histogram findings, with increased edema at the site of second impact in rmTBI 3d animals and elevated blood deposition in the rmTBI 7d group at the site of the first injury. Histological measurements revealed spatial overlap of regions containing blood deposition and microglial activation within the cortices of all animals. In conclusion, our findings suggest that there is a window of tissue vulnerability where a second distant mTBI, induced 7d after an initial injury, exacerbates tissue abnormalities consistent with hemorrhagic progression.
Collapse
|
32
|
Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol 2012; 11:720-31. [PMID: 22698888 DOI: 10.1016/s1474-4422(12)70104-7] [Citation(s) in RCA: 956] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intracerebral haemorrhage accounts for about 10-15% of all strokes and is associated with high mortality and morbidity. No successful phase 3 clinical trials for this disorder have been completed. In the past 6 years, the number of preclinical and clinical studies focused on intracerebral haemorrhage has risen. Important advances have been made in animal models of this disorder and in our understanding of mechanisms underlying brain injury after haemorrhage. Several therapeutic targets have subsequently been identified that are now being pursued in clinical trials. Many clinical trials have been based on limited preclinical data, and guidelines to justify taking preclinical results to the clinic are needed.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
33
|
Abstract
The likelihood of translating therapeutic interventions for stroke rests on the quality of preclinical science. Given the limited success of putative treatments for ischemic stroke and the reasons put forth to explain it, we sought to determine whether such problems hamper progress for intracerebral hemorrhage (ICH). Approximately 10% to 20% of strokes result from an ICH, which results in considerable disability and high mortality. Several animal models reproduce ICH and its underlying pathophysiology, and these models have been widely used to evaluate treatments. As yet, however, none has successfully translated. In this review, we focus on rodent models of ICH, highlighting differences among them (e.g., pathophysiology), issues with experimental design and analysis, and choice of end points. A Pub Med search for experimental ICH (years: 2007 to 31 July 2011) found 121 papers. Of these, 84% tested neuroprotectants, 11% tested stem cell therapies, and 5% tested rehabilitation therapies. We reviewed these to examine study quality (e.g., use of blinding procedures) and choice of end points (e.g., behavioral testing). Not surprisingly, the problems that have plagued the ischemia field are also prevalent in ICH literature. Based on these data, several recommendations are put forth to facilitate progress in identifying effective treatments for ICH.
Collapse
|
34
|
Abstract
Hypoalbuminemia is common among critically ill/injured patients and is strongly associated with increased morbidity and mortality in the patients with and without neurological conditions. Normal serum albumin is important as the primary intravascular antioxidant, in transporting a variety of hormones, medications and electrolytes, in providing colloid osmotic pressure during trans-compartmental fluid movement, in enhancing organ and tissue blood flow, and in supporting acid-base balance. Studies of albumin administration during intravascular resuscitation have not addressed potential longer term benefits to sustaining serum albumin concentrations during critical care. Evidence for such benefit is present although additional prospective studies are needed.
Collapse
Affiliation(s)
- David J Powner
- Departments of Neurosurgery and Internal Medicine, University of Texas Health Sciences Center at Houston Medical School, 6431 Fannin Street, MSB 7.154, Houston, TX 77030, USA.
| |
Collapse
|
35
|
McAuley G, Schrag M, Barnes S, Obenaus A, Dickson A, Kirsch W. In vivo iron quantification in collagenase-induced microbleeds in rat brain. Magn Reson Med 2011; 67:711-7. [PMID: 21721041 DOI: 10.1002/mrm.23045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/18/2011] [Accepted: 05/21/2011] [Indexed: 01/02/2023]
Abstract
Brain microbleeds (BMB) are associated with chronic and acute cerebrovascular disease. Because BMB present in the brain is a source of potentially cytotoxic iron proportional to the volume of extravasated blood, BMB iron content is a potentially valuable biomarker both to assess tissue risk and small cerebral vessel health. We recently reported methods to quantify focal iron sources using phase images that were tested in phantoms and BMB in postmortem tissue. In this study, we applied our methods to small hemorrhagic lesions induced in the in vivo rat brain using bacterial collagenase. As expected by theory, measurements of geometric features in phase images correlated with lesion iron content measured by graphite furnace atomic absorption spectrometry. Iron content estimation following BMB in an in vivo rodent model could shed light on the role and temporal evolution of iron-mediated tissue damage and efficacy of potential treatments in cerebrovascular diseases associated with BMB.
Collapse
Affiliation(s)
- Grant McAuley
- Neurosurgery Center for Research, Training and Education, Loma Linda University, Loma Linda, California 92354, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Intracerebral hemorrhage (ICH) imparts a higher mortality and morbidity than ischemic stroke. The therapeutic interventions that are currently available focus mainly on supportive care and secondary prevention. There is a paucity of evidence to support any one acute intervention that improves functional outcome. This chapter highlights current treatment targets for ICH based on the pathophysiology of the disease.
Collapse
Affiliation(s)
- Navdeep Sangha
- Department of Neurology, University of Texas Medical School-UT Health, 6431 Fannin, MSB 7.118, Houston, TX 77030 USA
| | - Nicole R. Gonzales
- Department of Neurology, University of Texas Medical School-UT Health, 6431 Fannin, MSB 7.118, Houston, TX 77030 USA
| |
Collapse
|
37
|
Frantzias J, Sena ES, Macleod MR, Al-Shahi Salman R. Treatment of intracerebral hemorrhage in animal models: meta-analysis. Ann Neurol 2011; 69:389-99. [PMID: 21387381 DOI: 10.1002/ana.22243] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Interventions that improve functional outcome after acute intracerebral hemorrhage (ICH) in animals might benefit humans. Therefore, we systematically reviewed the literature to find studies of nonsurgical treatments tested in animal models of ICH. METHODS In July 2009 we searched Ovid Medline (from 1950), Embase (from 1980), and ISI Web of Knowledge (from 1969) for controlled animal studies of nonsurgical interventions given after the induction of ICH that reported neurobehavioral outcome. We assessed study quality and performed meta-analysis using a weighted mean difference random effects model. RESULTS Of 13,343 publications, 88 controlled studies described the effects of 64 different medical interventions (given a median of 2 hours after ICH induction) on 38 different neurobehavioral scales in 2,616 treated or control animals (median 14 rodents per study). Twenty-seven (31%) studies randomized treatment allocation, and 7 (8%) reported allocation concealment; these studies had significantly smaller effect sizes than those without these attributes (p < 0.001). Of 64 interventions stem cells, calcium channel blockers, anti-inflammatory drugs, iron chelators, and estrogens improved both structural outcomes and neurobehavioral scores in >1 study. Meta-regression revealed that together, structural outcome and the intervention used accounted for 65% of the observed heterogeneity in neurobehavioral score (p < 0.001, adjusted r(2) = 0.65). INTERPRETATION Further animal studies of the interventions that we found to improve both functional and structural outcomes in animals, using better experimental designs, could target efforts to translate effective treatments for ICH in animals into randomized controlled trials in humans.
Collapse
Affiliation(s)
- Joseph Frantzias
- Division of Clinical Neurosciences, Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom
| | | | | | | |
Collapse
|
38
|
Obenaus A, Dilmac N, Tone B, Tian HR, Hartman R, Digicaylioglu M, Snyder EY, Ashwal S. Long-term magnetic resonance imaging of stem cells in neonatal ischemic injury. Ann Neurol 2010; 69:282-91. [PMID: 21387373 DOI: 10.1002/ana.22168] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 07/03/2010] [Accepted: 07/16/2010] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Quantitative magnetic resonance imaging (MRI) can serially and noninvasively assess the degree of injury in rat pup models of hypoxic ischemic injury (HII). It can also noninvasively monitor stem cell migration following iron oxide prelabeling. Reports have shown that neural stem cells (NSCs) may help mediate neuroprotection or stimulate neuroreparative responses in adult and neonatal models of ischemic injury. We investigated the ability of high-field MRI to monitor and noninvasively quantify the migration, proliferation, and location of iron oxide-labeled NSCs over very long time periods (58 weeks) in real time while contemporaneously correlating this activity with the evolving severity and extent of neural damage. METHODS Labeled clonal murine NSCs (mNSCs) were implanted 3 days after unilateral HII in 10-day-old rat pups into the contralateral striatum or ventricle. We developed methods for objectively quantifying key aspects of dynamic NSC behavior (eg, viability; extent, and speed of migration; degree of proliferation; extent of integration into host parenchyma). MRI images were validated with histological and immunohistochemical assessments. RESULTS mNSCs rapidly migrated (100 μm/day) to the lesion site. Chains of migrating NSCs were observed in the corpus callosum. In pups subjected to HII, though not in intact control animals, we observed a 273% increase in the MR-derived volume of mNSCs 4 weeks after implantation (correlating with the known proliferative behavior of endogenous and exogenous NSCs) that slowly declined over the 58-week time course, with no adverse consequences. Large numbers of now quiescent mNSCs remained at the site of injury, many retaining their iron oxide label. INTERPRETATION Our studies demonstrate that MRI can simultaneously monitor evolving neonatal cerebral injury as well as NSC migration and location. Most importantly, it can noninvasively monitor proliferation dynamically for prolonged time periods. To be able to pursue clinical trials in newborns using stem cell therapies it is axiomatic that safety be insured through the long-term real time monitoring of cell fate and activity, particularly with regard to observing unanticipated risks to the developing brain. This study supports the feasibility of reliably using MRI for this purpose.
Collapse
Affiliation(s)
- Andre Obenaus
- Department Radiation Medicine, Loma Linda University, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Beray-Berthat V, Delifer C, Besson VC, Girgis H, Coqueran B, Plotkine M, Marchand-Leroux C, Margaill I. Long-term histological and behavioural characterisation of a collagenase-induced model of intracerebral haemorrhage in rats. J Neurosci Methods 2010; 191:180-90. [DOI: 10.1016/j.jneumeth.2010.06.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 06/19/2010] [Indexed: 10/19/2022]
|
40
|
Abstract
Molecular imaging provides spatial and temporal information on cellular changes that occur during development and in disease. MRI and optical imaging of reporter genes allows for the visualization of promoter activity, protein-protein interactions, protein stability and the tracking of individual proteins and cells. Reporter genes can be genetically encoded in transgenic animals or detected through the administration of an exogenous contrast agent. Advances in molecular imaging of reporter genes have led to the development of imaging probes that detect changes in endogenous cellular changes. The ability to use contrast agents coupled with functional information on cellular events will allow for sensitive assessment of individual patient therapies, leading to an accurately tailored treatment regimen.
Collapse
Affiliation(s)
- Allison S. Harney
- Departments of Chemistry, Biochemistry and Molecular and Cell Biology, Neurobiology and Physiology, and Radiology, Northwestern University, Evanston, IL, 60208, USA
| | - Thomas J. Meade
- Departments of Chemistry, Biochemistry and Molecular and Cell Biology, Neurobiology and Physiology, and Radiology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
41
|
Titova E, Adami A, Ostrowski R, Lalas S, Vlkolinsky R, Zhang JH, Nelson G, Obenaus A. Radiation exposure prior to ischemia decreases lesion volume, brain edema and cell death. ACTA NEUROCHIRURGICA. SUPPLEMENT 2010; 106:51-3. [PMID: 19812920 DOI: 10.1007/978-3-211-98811-4_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE To investigate the neuronal response to ischemic injury following exposure to whole brain proton irradiation. METHODS Brain only proton irradiation (8 Gy, 250 MeV) was performed ten days prior to middle cerebral artery occlusion (MCAO) in 1 year old male Sprague Dawley rats. MCAO was induced in two animal groups: proton irradiated (MCAO + Rad) and MCAO only. Magnetic resonance imaging (MRI) and quantitative analysis were performed prior to and 2 days after irradiation, and then 2, 14 and 28 days after MCAO. After the last imaging time point animals were sacrificed and TUNEL staining was performed on 4% paraformaldehyde - fixed brain sections. RESULTS Neuroimaging demonstrated a reduction in ischemic lesion volume in the MCAO + Rad group compared with MCAO alone. Neurological deficits did not differ between ischemia groups. Interestingly, there was a 34% decrease in the number of TUNEL-positive cells in MCAO + Rad brains compared to MCAO alone. CONCLUSION Our results suggest that radiation treatment reduces brain edema, ischemic lesion volume and peri-ischemic apoptosis. The underlying mechanisms are currently unknown and additional studies will elucidate the significance of these results.
Collapse
Affiliation(s)
- E Titova
- Department of Radiation Medicine, Loma Linda University, Chan Shun Pavilion, Loma Linda, CA 92350, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Adeleye A, Shohami E, Nachman D, Alexandrovich A, Trembovler V, Yaka R, Shoshan Y, Dhawan J, Biegon A. D-cycloserine improves functional outcome after traumatic brain injury with wide therapeutic window. Eur J Pharmacol 2009; 629:25-30. [PMID: 19958766 DOI: 10.1016/j.ejphar.2009.11.066] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 11/15/2009] [Accepted: 11/24/2009] [Indexed: 02/02/2023]
Abstract
It has been long thought that hyperactivation of N-methyl-D-aspartate (NMDA) receptors underlies neurological decline after traumatic brain injury. However, all clinical trials with NMDA receptor antagonists failed. Since NMDA receptors are down-regulated from 4h to 2weeks after brain injury, activation at 24h, rather than inhibition, of these receptors, was previously shown to be beneficial in mice. Here, we tested the therapeutic window, dose regimen and mechanism of action of the NMDA receptor partial agonist D-cycloserine (DCS) in traumatic brain injury. Male mice were subjected to trauma using a weight-drop model, and administered 10mg/kg (i.p.) DCS or vehicle once (8, 16, 24, or 72h) twice (24 and 48h) or three times (24, 48 and 72h). Functional recovery was assessed for up to 60days, using a Neurological Severity Score that measures neurobehavioral parameters. In all groups in which treatment was begun at 24 or 72h neurobehavioral function was significantly better than in the vehicle-treated groups. Additional doses, on days 2 and 3 did not further improve recovery. Mice treated at 8h or 16h post injury did not differ from the vehicle-treated controls. Co-administration of the NMDA receptor antagonist MK-801 completely blocked the protective effect of DCS given at 24h. Infarct volume measured by 2,3,5-triphenyltetrazolium chloride staining at 48h or by cresyl violet at 28days was not affected by DCS treatment. Since DCS is used clinically for other indications, the present study offers a novel approach for treating human traumatic brain injury with a therapeutic window of at least 24h.
Collapse
Affiliation(s)
- Amos Adeleye
- Department of Pharmacology, The Hebrew University School of Pharmacy, Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Karki K, Knight RA, Han Y, Yang D, Zhang J, Ledbetter KA, Chopp M, Seyfried DM. Simvastatin and atorvastatin improve neurological outcome after experimental intracerebral hemorrhage. Stroke 2009; 40:3384-9. [PMID: 19644071 DOI: 10.1161/strokeaha.108.544395] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE This study investigates the effects of statin treatment on experimental intracerebral hemorrhage (ICH) using behavioral, histological, and MRI measures of recovery. METHODS Primary ICH was induced in rats. Simvastatin (2 mg/kg), atorvastatin (2 mg/kg), or phosphate-buffered saline (n=6 per group) was given daily for 1 week. MRI studies were performed 2 to 3 days before ICH, and at 1 to 2 hours and 1, 2, 7, 14, and 28 days after ICH. The ICH evolution was assessed via hematoma volume measurements using susceptibility-weighted imaging (SWI) and tissue loss using T2 maps and hematoxylin and eosin (H&E) histology. Neurobehavioral tests were done before ICH and at various time points post-ICH. Additional histological measures were performed with doublecortin neuronal nuclei and bromodeoxyuridine stainings. RESULTS Initial ICH volumes determined by SWI were similar across all groups. Simvastatin significantly reduced hematoma volume at 4 weeks (P=0.002 versus control with acute volumes as baseline), whereas that for atorvastatin was marginal (P=0.09). MRI estimates of tissue loss (% of contralateral hemisphere) for treated rats were significantly lower (P=0.0003 and 0.001, respectively) than for control at 4 weeks. Similar results were obtained for H&E histology (P=0.0003 and 0.02, respectively). Tissue loss estimates between MRI and histology were well correlated (R2=0.764, P<0.0001). Significant improvement in neurological function was seen 2 to 4 weeks post-ICH with increased neurogenesis observed. CONCLUSIONS Simvastatin and atorvastatin significantly improved neurological recovery, decreased tissue loss, and increased neurogenesis when administered for 1 week after ICH.
Collapse
Affiliation(s)
- Kishor Karki
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
MacLellan CL, Langdon KD, Churchill KP, Granter-Button S, Corbett D. Assessing cognitive function after intracerebral hemorrhage in rats. Behav Brain Res 2008; 198:321-8. [PMID: 19041895 DOI: 10.1016/j.bbr.2008.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 10/24/2008] [Accepted: 11/03/2008] [Indexed: 01/27/2023]
Abstract
Preclinical studies must rigorously assess whether putative therapies improve motor and cognitive function following brain injury. Intracerebral hemorrhage (ICH) causes significant sensory-motor and cognitive deficits in humans. However, no study has evaluated cognition in rodent ICH models. Thus, we used a battery of tests to comprehensively examine whether a striatal ICH causes cognitive impairments in rats. Bacterial collagenase (or sterile saline for SHAM surgery) was injected into the striatum to create an ICH. Two days later, functional deficits were assessed using a neurological deficit scale (NDS), which is most sensitive to ICH injury. Sensory and/or motor deficits may confound cognitive testing; thus, we waited until these had resolved before testing learning and memory. Testing was conducted 1-7 months after ICH and included spontaneous alternation, elevated plus maze, open-field, Morris water maze, T-maze (win-shift and win-stay paradigms), and the radial arm maze (eight and four arms baited protocols). Significant motor deficits at 2 days completely resolved by 1 month, at which time cognitive testing began. In contrast to persistent cognitive deficits that occur after ICH in humans, we did not detect significant learning or memory deficits after ICH in rats. Our results suggest that these tests will not likely be useful for assessing outcome in experimental ICH studies. In conclusion, animal models that better mimic clinical ICH (both motor and cognitive deficits) must be developed. This may include increasing ICH severity or injuring other functional subdivisions within the striatum that may lead to more profound cognitive deficits.
Collapse
Affiliation(s)
- Crystal L MacLellan
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada A1B 3V6.
| | | | | | | | | |
Collapse
|
45
|
Knight RA, Han Y, Nagaraja TN, Whitton P, Ding J, Chopp M, Seyfried DM. Temporal MRI assessment of intracerebral hemorrhage in rats. Stroke 2008; 39:2596-602. [PMID: 18635862 DOI: 10.1161/strokeaha.107.506683] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE MRI was used to evaluate the effects of experimental intracerebral hemorrhage (ICH) on brain tissue injury and recovery. METHODS Primary ICH was induced in rats (n=6) by direct infusion of autologous blood into the striatum. The evolution of ICH damage was assessed by MRI estimates of T(2) and T(1sat) relaxation times, cerebral blood flow, vascular permeability, and susceptibility-weighted imaging before surgery (baseline) and at 2 hours and 1, 7, and 14 days post-ICH. Behavioral testing was done before and at 1, 7, and 14 days post-ICH. Animals were euthanized for histology at 14 days. RESULTS The MRI appearance of the hemorrhage and surrounding regions changed in a consistent manner over time. Two primary regions of interest were identified based on T(2) values. These included a core, corresponding to the bulk of the hemorrhage, and an adjacent rim; both varied with time. The core was associated with significantly lower cerebral blood flow values at all post-ICH time points, whereas cerebral blood flow varied in the rim. Increases in vascular permeability were noted at 1, 7, and 14 days. Changes in T(1sat) were similar to those of T(2). MRI and histological estimates of tissue loss were well correlated and showed approximately 9% hemispheric tissue loss. CONCLUSIONS Although the cerebral blood flow changes observed with this ICH model may not exactly mimic the clinical situation, our results suggest that the evolution of ICH injury can be accurately characterized with MRI. These methods may be useful to evaluate therapeutic interventions after experimental ICH and eventually in humans.
Collapse
Affiliation(s)
- Robert A Knight
- Henry Ford Hospital, Department of Neurology-NMR Research, 2799 West Grand Blvd, Detroit, MI 48202, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Kuluz J, Huang T, Watson B, Vannucci S. Stroke in the immature brain: review of pathophysiology and animal models of pediatric stroke. FUTURE NEUROLOGY 2008. [DOI: 10.2217/14796708.3.2.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pediatric stroke research presents many challenges. Relatively low incidence, need for age stratification, diverse etiologies, delays in diagnosis, lack of an established age-based stroke severity scale and outcome measures are only some of the issues that have prevented the implementation of clinical trials in infants and children with stroke. Experimental animal models of pediatric stroke, therefore, are critical to understanding the pathophysiology and management of ischemic brain damage in the immature brain, and provide the necessary platform for future clinical trials. In this review we discuss the pertinent clinical aspects of pediatric stroke, the pathophysiology of stroke in the developing brain and the animal models established to study basic mechanisms as well as translational issues in pediatric stroke.
Collapse
Affiliation(s)
- John Kuluz
- Associate Professor of Pediatrics, University of Miami, Department of Pediatrics (R-131), Miller School of Medicine, PO Box 016960, Miami, FL 33101, USA
| | - Tingting Huang
- Post-Doctoral Research Associate, University of Miami, Department of Pediatrics (R-131), Miller School of Medicine, PO Box 016960 Miami, FL 33101, USA
| | - Brant Watson
- Professor of Neurology, University of Miami, Department of Neurology (D4–5), Miller School of Medicine, PO Box 016960, Miami, FL 33136, USA
| | - Susan Vannucci
- Research Professor of Neuroscience in Pediatrics/Newborn Medicine, Weill Cornell Medical College, 525 East 68th Street, N-506, NY 10065, USA
| |
Collapse
|
47
|
The effects of selective brain hypothermia on intracerebral hemorrhage in rats. Exp Neurol 2007; 208:277-84. [DOI: 10.1016/j.expneurol.2007.08.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 08/23/2007] [Accepted: 08/28/2007] [Indexed: 11/20/2022]
|