1
|
Hu Z, Deng X, Zhou S, Zhou C, Shen M, Gao X, Huang Y. Pathogenic mechanisms and therapeutic implications of extracellular matrix remodelling in cerebral vasospasm. Fluids Barriers CNS 2023; 20:81. [PMID: 37925414 PMCID: PMC10625254 DOI: 10.1186/s12987-023-00483-8] [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: 06/14/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
Cerebral vasospasm significantly contributes to poor prognosis and mortality in patients with aneurysmal subarachnoid hemorrhage. Current research indicates that the pathological and physiological mechanisms of cerebral vasospasm may be attributed to the exposure of blood vessels to toxic substances, such as oxyhaemoglobin and inflammation factors. These factors disrupt cerebral vascular homeostasis. Vascular homeostasis is maintained by the extracellular matrix (ECM) and related cell surface receptors, such as integrins, characterised by collagen deposition, collagen crosslinking, and elastin degradation within the vascular ECM. It involves interactions between the ECM and smooth muscle cells as well as endothelial cells. Its biological activities are particularly crucial in the context of cerebral vasospasm. Therefore, regulating ECM homeostasis may represent a novel therapeutic target for cerebral vasospasm. This review explores the potential pathogenic mechanisms of cerebral vasospasm and the impacts of ECM protein metabolism on the vascular wall during ECM remodelling. Additionally, we underscore the significance of an ECM protein imbalance, which can lead to increased ECM stiffness and activation of the YAP pathway, resulting in vascular remodelling. Lastly, we discuss future research directions.
Collapse
Affiliation(s)
- Ziliang Hu
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Liuting Street 59, Ningbo, 315010, Zhejiang, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, 315302, Zhejiang, China
| | - Xinpeng Deng
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Liuting Street 59, Ningbo, 315010, Zhejiang, China
| | - Shengjun Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Liuting Street 59, Ningbo, 315010, Zhejiang, China
| | - Chenhui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Liuting Street 59, Ningbo, 315010, Zhejiang, China
| | - Menglu Shen
- Cixi Third People's Hospital, Cixi, 315324, Zhejiang, China
| | - Xiang Gao
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Liuting Street 59, Ningbo, 315010, Zhejiang, China.
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Liuting Street 59, Ningbo, 315010, Zhejiang, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
2
|
Becker K. Animal Welfare Aspects in Planning and Conducting Experiments on Rodent Models of Subarachnoid Hemorrhage. Cell Mol Neurobiol 2023; 43:3965-3981. [PMID: 37861870 DOI: 10.1007/s10571-023-01418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
Subarachnoid hemorrhage is an acute life-threatening cerebrovascular disease with high socio-economic impact. The most frequent cause, the rupture of an intracerebral aneurysm, is accompanied by abrupt changes in intracerebral pressure, cerebral perfusion pressure and, consequently, cerebral blood flow. As aneurysms rupture spontaneously, monitoring of these parameters in patients is only possible with a time delay, upon hospitalization. To study alterations in cerebral perfusion immediately upon ictus, animal models are mandatory. This article addresses the points necessarily to be included in an animal project proposal according to EU directive 2010/63/EU for the protection of animals used for scientific purposes and herewith offers an insight into animal welfare aspects of using rodent models for the investigation of cerebral perfusion after subarachnoid hemorrhage. It compares surgeries, model characteristics, advantages, and drawbacks of the most-frequently used rodent models-the endovascular perforation model and the prechiasmatic and single or double cisterna magna injection model. The topics of discussing anesthesia, advice on peri- and postanesthetic handling of animals, assessing the severity of suffering the animals undergo during the procedure according to EU directive 2010/63/EU and weighing the use of these in vivo models for experimental research ethically are also presented. In conclusion, rodent models of subarachnoid hemorrhage display pathophysiological characteristics, including changes of cerebral perfusion similar to the clinical situation, rendering the models suited to study the sequelae of the bleeding. A current problem is low standardization of the models, wherefore reporting according to the ARRIVE guidelines is highly recommended. Animal welfare aspects of rodent models of subarachnoid hemorrhage. Rodent models for investigation of cerebral perfusion after subarachnoid hemorrhage are compared regarding surgeries and model characteristics, and 3R measures are suggested. Anesthesia is discussed, and advice given on peri- and postanesthetic handling. Severity of suffering according to 2010/63/EU is assessed and use of these in vivo models weighed ethically.
Collapse
Affiliation(s)
- Katrin Becker
- Institute for Translational Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
- Institute for Cardiovascular Sciences, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
3
|
Tartara F, Montalbetti A, Crobeddu E, Armocida D, Tavazzi E, Cardia A, Cenzato M, Boeris D, Garbossa D, Cofano F. Compartmental Cerebrospinal Fluid Events Occurring after Subarachnoid Hemorrhage: An "Heparin Oriented" Systematic Review. Int J Mol Sci 2023; 24:7832. [PMID: 37175544 PMCID: PMC10178276 DOI: 10.3390/ijms24097832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) represents a severe acute event with high morbidity and mortality due to the development of early brain injury (EBI), secondary delayed cerebral ischemia (DCI), and shunt-related hydrocephalus. Secondary events (SSE) such as neuroinflammation, vasospasm, excitotoxicity, blood-brain barrier disruption, oxidative cascade, and neuronal apoptosis are related to DCI. Despite improvement in management strategies and therapeutic protocols, surviving patients frequently present neurological deficits with neurocognitive impairment. The aim of this paper is to offer to clinicians a practical review of the actually documented pathophysiological events following subarachnoid hemorrhage. To reach our goal we performed a literature review analyzing reported studies regarding the mediators involved in the pathophysiological events following SAH occurring in the cerebrospinal fluid (CSF) (hemoglobin degradation products, platelets, complement, cytokines, chemokines, leucocytes, endothelin-1, NO-synthase, osteopontin, matricellular proteins, blood-brain barrier disruption, microglia polarization). The cascade of pathophysiological events secondary to SAH is very complex and involves several interconnected, but also distinct pathways. The identification of single therapeutical targets or specific pharmacological agents may be a limited strategy able to block only selective pathophysiological paths, but not the global evolution of SAH-related events. We report furthermore on the role of heparin in SAH management and discuss the rationale for use of intrathecal heparin as a pleiotropic therapeutical agent. The combination of the anticoagulant effect and the ability to interfere with SSE theoretically make heparin a very interesting molecule for SAH management.
Collapse
Affiliation(s)
- Fulvio Tartara
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Montalbetti
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Emanuela Crobeddu
- A.O.U. Maggiore della Carità University Hospital, Department of Neurosurgery, 28100 Novara, Italy
| | - Daniele Armocida
- A.U.O. Policlinico Umberto I, Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00185 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Eleonora Tavazzi
- IRCCS Fondazione Istituto Neurologico Nazionale C. Mondino, 27100 Pavia, Italy
| | - Andrea Cardia
- Department of Neurosurgery, Neurocenter of Southern Switzerland, EOC, 6900 Lugano, Switzerland
| | - Marco Cenzato
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Davide Boeris
- Ospedale Niguarda Ca’ Granda, Department of Neurosurgery, 20162 Milan, Italy
| | - Diego Garbossa
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| | - Fabio Cofano
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10095 Turin, Italy
| |
Collapse
|
4
|
The protective effect of low-dose minocycline on brain microvascular ultrastructure in a rodent model of subarachnoid hemorrhage. Histochem Cell Biol 2023; 159:91-114. [PMID: 36153470 PMCID: PMC9899762 DOI: 10.1007/s00418-022-02150-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 02/07/2023]
Abstract
The multifaceted nature of subarachnoid hemorrhage (SAH) pathogenesis is poorly understood. To date, no pharmacological agent has been found to be efficacious for the prevention of brain injury when used for acute SAH intervention. This study was undertaken to evaluate the beneficial effects of low-dose neuroprotective agent minocycline on brain microvascular ultrastructures that have not been studied in detail. We studied SAH brain injury using an in vivo prechiasmatic subarachnoid hemorrhage rodent model. We analyzed the qualitative and quantitative ultrastructural morphology of capillaries and surrounding neuropil in the rodent brains with SAH and/or minocycline administration. Here, we report that low-dose minocycline (1 mg/kg) displayed protective effects on capillaries and surrounding cells from significant SAH-induced changes. Ultrastructural morphology analysis revealed also that minocycline stopped endothelial cells from abnormal production of vacuoles and vesicles that compromise blood-brain barrier (BBB) transcellular transport. The reported ultrastructural abnormalities as well as neuroprotective effects of minocycline during SAH were not directly mediated by inhibition of MMP-2, MMP-9, or EMMPRIN. However, SAH brain tissue treated with minocycline was protected from development of other morphological features associated with oxidative stress and the presence of immune cells in the perivascular space. These data advance the knowledge on the effect of SAH on brain tissue ultrastructure in an SAH rodent model and the neuroprotective effect of minocycline when administered in low doses.
Collapse
|
5
|
Brain Oxygen-Directed Management of Aneurysmal Subarachnoid Hemorrhage. Temporal Patterns of Cerebral Ischemia During Acute Brain Attack, Early Brain Injury, and Territorial Sonographic Vasospasm. World Neurosurg 2022; 166:e215-e236. [PMID: 35803565 DOI: 10.1016/j.wneu.2022.06.149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neurocritical management of aneurysmal subarachnoid hemorrhage focuses on delayed cerebral ischemia (DCI) after aneurysm repair. METHODS This study conceptualizes the pathophysiology of cerebral ischemia and its management using a brain oxygen-directed protocol (intracranial pressure [ICP] control, eubaric hyperoxia, hemodynamic therapy, arterial vasodilation, and neuroprotection) in patients with subarachnoid hemorrhage, undergoing aneurysm clipping (n = 40). RESULTS The brain oxygen-directed protocol reduced Lbo2 (Pbto2 [partial pressure of brain tissue oxygen] <20 mm Hg) from 67% to 15% during acute brain attack (<24 hours of ictus), by increasing Pbto2 from 11.31 ± 9.34 to 27.85 ± 6.76 (P < 0.0001) and then to 29.09 ± 17.88 within 72 hours. Day-after-bleed, Fio2 change, ICP, hemoglobin, and oxygen saturation were predictors for Pbto2 during early brain injury. Transcranial Doppler ultrasonography velocities (>20 cm/second) increased at day 2. During DCI caused by territorial sonographic vasospasm (TSV), middle cerebral artery mean velocity (Vm) increased from 45.00 ± 15.12 to 80.37 ± 38.33/second by day 4 with concomitant Pbto2 reduction from 29.09 ± 17.88 to 22.66 ± 8.19. Peak TSV (days 7-12) coincided with decline in Pbto2. Nicardipine mitigated Lbo2 during peak TSV, in contrast to nimodipine, with survival benefit (P < 0.01). Intravenous and cisternal nicardipine combination had survival benefit (Cramer Φ = 0.43 and 0.327; G2 = 28.32; P < 0.001). This study identifies 4 zones of Lbo2 during survival benefit (Cramer Φ = 0.43 and 0.3) TSV, uncompensated; global cerebral ischemia, compensated, and normal Pbto2. Admission Glasgow Coma Scale score (not increased ICP) was predictive of low Pbto2 (β = 0.812, R2 = 0.661, F1,30 = 58.41; P < 0.0001) during early brain injury. Coma was the only credible predictor for mortality (odds ratio, 7.33/>4.8∗; χ2 = 7.556; confidence interval, 1.70-31.54; P < 0.01) followed by basilar aneurysm, poor grade, high ICP and Lbo2 during TSV. Global cerebral ischemia occurs immediately after the ictus, persisting in 30% of patients despite the high therapeutic intensity level, superimposed by DCI during TSV. CONCLUSIONS We propose implications for clinical practice and patient management to minimize cerebral ischemia.
Collapse
|
6
|
Feng D, Zhou J, Liu H, Wu X, Li F, Zhao J, Zhang Y, Wang L, Chao M, Wang Q, Qin H, Ge S, Liu Q, Zhang J, Qu Y. Astrocytic NDRG2-PPM1A interaction exacerbates blood-brain barrier disruption after subarachnoid hemorrhage. SCIENCE ADVANCES 2022; 8:eabq2423. [PMID: 36179025 PMCID: PMC9524825 DOI: 10.1126/sciadv.abq2423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/15/2022] [Indexed: 06/01/2023]
Abstract
Blood-brain barrier (BBB) injury critically exacerbates the poor prognosis of patients with subarachnoid hemorrhage (SAH). The massively increased matrix metalloproteinases 9 (MMP-9) plays a deleterious role in BBB. However, the main source and mechanism of MMP-9 production after SAH remain unclear. We reported that the increased MMP-9 was mainly derived from reactive astrocytes after SAH. Ndrg2 knockout in astrocytes inhibited MMP-9 expression after SAH and attenuated BBB damage. Astrocytic Ndrg2 knockout decreased the phosphorylation of Smad2/3 and the transcription of MMP-9. Notably, cytoplasmic NDRG2 bound to the protein phosphatase PPM1A and restricted the dephosphorylation of Smad2/3. Accordingly, TAT-QFNP12, a novel engineered peptide that could block the NDRG2-PPM1A binding and reduce Smad2/3 dephosphorylation, decreased astrocytic MMP-9 production and BBB disruption after SAH. In conclusion, this study identified NDRG2-PPM1A signaling in reactive astrocytes as a key switch for MMP-9 production and provided a novel therapeutic avenue for BBB protection after SAH.
Collapse
Affiliation(s)
- Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Jinpeng Zhou
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Xun Wu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Fei Li
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Junlong Zhao
- Department of Medical Genetics and Development Biology, Fourth Military Medical University, Xi’an 710032, China
| | - Yu Zhang
- Department of Biological Sciences, Xinyang Normal University, Xinyang 464000, China
| | - Lei Wang
- Department of Biological Sciences, Xinyang Normal University, Xinyang 464000, China
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Qiang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Huaizhou Qin
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jian Zhang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi’an 710032, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, International Cooperation Platform for Encephalopathy of Shaanxi Province, Xi’an 710038, China
| |
Collapse
|
7
|
Strickland BA, Barisano G, Abedi A, Shiroishi MS, Cen S, Emanuel B, Bulic S, Kim-Tenser M, Nguyen P, Giannotta SL, Mack W, Russin J. Minocycline decreases blood-brain barrier permeability following aneurysmal subarachnoid hemorrhage: a randomized, double-blind, controlled trial. J Neurosurg 2022; 136:1251-1259. [PMID: 35349976 DOI: 10.3171/2021.6.jns211270] [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: 05/20/2021] [Accepted: 06/18/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Aneurysmal subarachnoid hemorrhage (aSAH)-induced vasospasm is linked to increased inflammatory cell trafficking across a permeable blood-brain barrier (BBB). Elevations in serum levels of matrix metalloprotease 9 (MMP9), a BBB structural protein, have been implicated in the pathogenesis of vasospasm onset. Minocycline is a potent inhibitor of MMP9. The authors sought to detect an effect of minocycline on BBB permeability following aSAH. METHODS Patients presenting within 24 hours of symptom onset with imaging confirmed aSAH (Fisher grade 3 or 4) were randomized to high-dose (10 mg/kg) minocycline or placebo. The primary outcome of interest was BBB permeability as quantitated by contrast signal intensity ratios in vascular regions of interest on postbleed day (PBD) 5 magnetic resonance permeability imaging. Secondary outcomes included serum MMP9 levels and radiographic and clinical evidence of vasospasm. RESULTS A total of 11 patients were randomized to minocycline (n = 6) or control (n = 5) groups. No adverse events or complications attributable to minocycline were reported. High-dose minocycline administration was associated with significantly lower permeability indices on imaging analysis (p < 0.01). There was no significant difference with respect to serum MMP9 levels between groups, although concentrations trended upward in both cohorts. Radiographic vasospasm was noted in 6 patients (minocycline = 3, control = 3), with only 1 patient developing symptoms of clinical vasospasm in the minocycline cohort. There was no difference between cohorts with respect to Lindegaard ratios, transcranial Doppler values, or onset of vasospasm. CONCLUSIONS Minocycline at high doses is well tolerated in the ruptured cerebral aneurysm population. Minocycline curtails breakdown of the BBB following aSAH as evidenced by lower permeability indices, though minocycline did not significantly alter serum MMP9 levels. Larger randomized clinical trials are needed to assess minocycline as a neuroprotectant against aSAH-induced vasospasm. Clinical trial registration no.: NCT04876638 (clinicaltrials.gov).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jonathan Russin
- 1Departments of Neurosurgery
- 5Neurorestoration Center, University of Southern California, Los Angeles, California
| |
Collapse
|
8
|
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS 2022; 19:29. [PMID: 35410231 PMCID: PMC8996682 DOI: 10.1186/s12987-022-00312-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The response of the blood-brain barrier (BBB) following a stroke, including subarachnoid hemorrhage (SAH), has been studied extensively. The main components of this reaction are endothelial cells, pericytes, and astrocytes that affect microglia, neurons, and vascular smooth muscle cells. SAH induces alterations in individual BBB cells, leading to brain homeostasis disruption. Recent experiments have uncovered many pathophysiological cascades affecting the BBB following SAH. Targeting some of these pathways is important for restoring brain function following SAH. BBB injury occurs immediately after SAH and has long-lasting consequences, but most changes in the pathophysiological cascades occur in the first few days following SAH. These changes determine the development of early brain injury as well as delayed cerebral ischemia. SAH-induced neuroprotection also plays an important role and weakens the negative impact of SAH. Supporting some of these beneficial cascades while attenuating the major pathophysiological pathways might be decisive in inhibiting the negative impact of bleeding in the subarachnoid space. In this review, we attempt a comprehensive overview of the current knowledge on the molecular and cellular changes in the BBB following SAH and their possible modulation by various drugs and substances.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Klaudia Lakatosová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
9
|
Okada T, Suzuki H, Travis ZD, Altay O, Tang J, Zhang JH. SPARC Aggravates Blood-Brain Barrier Disruption via Integrin αV β3/MAPKs/MMP-9 Signaling Pathway after Subarachnoid Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9739977. [PMID: 34804372 PMCID: PMC8601826 DOI: 10.1155/2021/9739977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/18/2021] [Indexed: 01/17/2023]
Abstract
Blood-brain barrier (BBB) disruption is a common and critical pathology following subarachnoid hemorrhage (SAH). We investigated the BBB disruption property of secreted protein acidic and rich in cysteine (SPARC) after SAH. A total of 197 rats underwent endovascular perforation to induce SAH or sham operation. Small interfering ribonucleic acid (siRNA) for SPARC or scrambled siRNA was administered intracerebroventricularly to rats 48 h before SAH. Anti-SPARC monoclonal antibody (mAb) 236 for functional blocking or normal mouse immunoglobulin G (IgG) was administered intracerebroventricularly 1 h after SAH. Selective integrin αVβ3 inhibitor cyclo(-RGDfK) or phosphate-buffered saline was administered intranasally 1 h before SAH, along with recombinant SPARC treatment. Neurobehavior, SAH severity, brain edema, immunohistochemical staining, and Western blot were evaluated. The expression of SPARC and integrin αVβ3 was upregulated after SAH in the endothelial cells. SPARC siRNA and anti-SPARC mAb 236 prevented neuroimpairments and brain edema through protection of BBB as measured by IgG extravasation 24 and 72 h after SAH. Recombinant SPARC aggravated neuroimpairments and cyclo(-RGDfK) suppressed the harmful neurological effects via inhibition of activated c-Jun N-terminal kinase, p38, and matrix metalloproteinase-9 followed by retention of endothelial junction proteins. SPARC may induce post-SAH BBB disruption via integrin αVβ3 signaling pathway.
Collapse
Affiliation(s)
- Takeshi Okada
- Department of Neurosurgery, Kuwana City Medical Center, 3-11 Kotobuki-cho, Kuwana, Mie 511-0061, Japan
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Zachary D. Travis
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
- Department of Earth and Biological Sciences, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
| | - Orhan Altay
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
- Department of Anesthesiology, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
- Department of Neurosurgery, Loma Linda University, Risley Hall, Room 219, 11041 Campus St., Loma Linda, CA 92354, USA
| |
Collapse
|
10
|
Stetter C, Weidner F, Lilla N, Weiland J, Kunze E, Ernestus RI, Muellenbach RM, Westermaier T. Therapeutic hypercapnia for prevention of secondary ischemia after severe subarachnoid hemorrhage: physiological responses to continuous hypercapnia. Sci Rep 2021; 11:11715. [PMID: 34083595 PMCID: PMC8175721 DOI: 10.1038/s41598-021-91007-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Temporary hypercapnia has been shown to increase cerebral blood flow (CBF) and might be used as a therapeutical tool in patients with severe subarachnoid hemorrhage (SAH). It was the aim of this study was to investigate the optimum duration of hypercapnia. This point is assumed to be the time at which buffer systems become active, cause an adaptation to changes of the arterial partial pressure of carbon dioxide (PaCO2) and annihilate a possible therapeutic effect. In this prospective interventional study in a neurosurgical ICU the arterial partial pressure of carbon dioxide (PaCO2) was increased to a target range of 55 mmHg for 120 min by modification of the respiratory minute volume (RMV) one time a day between day 4 and 14 in 12 mechanically ventilated poor-grade SAH-patients. Arterial blood gases were measured every 15 min. CBF and brain tissue oxygen saturation (StiO2) were the primary and secondary end points. Intracranial pressure (ICP) was controlled by an external ventricular drainage. Under continuous hypercapnia (PaCO2 of 53.17 ± 5.07), CBF was significantly elevated between 15 and 120 min after the start of hypercapnia. During the course of the trial intervention, cardiac output also increased significantly. To assess the direct effect of hypercapnia on brain perfusion, the increase of CBF was corrected by the parallel increase of cardiac output. The maximum direct CBF enhancing effect of hypercapnia of 32% was noted at 45 min after the start of hypercapnia. Thereafter, the CBF enhancing slowly declined. No relevant adverse effects were observed. CBF and StiO2 reproducibly increased by controlled hypercapnia in all patients. After 45 min, the curve of CBF enhancement showed an inflection point when corrected by cardiac output. It is concluded that 45 min might be the optimum duration for a therapeutic use and may provide an optimal balance between the benefits of hypercapnia and risks of a negative rebound effect after return to normal ventilation parameters. Trial registration: The study was approved by the institutional ethics committee (AZ 230/14) and registered at ClinicalTrials.gov (Trial-ID: NCT01799525). Registered 01/01/2015.
Collapse
Affiliation(s)
- Christian Stetter
- Department of Neurosurgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany.
| | - Franziska Weidner
- Department of Neurosurgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany.,Department of Neuroradiology, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - Judith Weiland
- Department of Neurosurgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Ekkehard Kunze
- Department of Neurosurgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| | - Ralf Michael Muellenbach
- Department of Anesthesia and Critical Care, University Hospital Wuerzburg, Oberduerrbacherstrasse 6, 97080, Wuerzburg, Germany.,Department of Anesthesiology, Klinikum Kassel, Moenchebergstrasse 41-43, 34125, Kassel, Germany
| | - Thomas Westermaier
- Department of Neurosurgery, University Hospital Wuerzburg, Josef-Schneider-Strasse 11, 97080, Wuerzburg, Germany
| |
Collapse
|
11
|
Amoo M, Henry J, Pender N, Brennan P, Campbell M, Javadpour M. Blood-brain barrier permeability imaging as a predictor for delayed cerebral ischaemia following subarachnoid haemorrhage. A narrative review. Acta Neurochir (Wien) 2021; 163:1457-1467. [PMID: 33404877 DOI: 10.1007/s00701-020-04670-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/01/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND Aneurysmal subarachnoid haemorrhage is associated with significant morbidity and mortality due to the myriad of complications contributing to early brain injury and delayed cerebral ischaemia. There is increasing interest in the exploration of the association between blood-brain barrier integrity and risks of delayed cerebral ischaemia and poor outcomes. Despite recent advances in cerebral imaging, radiographic imaging of blood-brain barrier disruption, as a biomarker for outcome prediction, has not been adopted in clinical practice. METHODS We performed a narrative review by searching for articles describing molecular changes or radiological identification of changes in BBB permeability following subarachnoid haemorrhage (SAH) on MEDLINE. Preclinical studies were analysed if reported structural changes and clinical studies were included if they investigated for radiological markers of BBB disruption and its correlation with delayed cerebral ischaemia. RESULTS There is ample preclinical evidence to suggest that there are structural changes in BBB permeability following SAH. The available clinical literature has demonstrated correlations between permeability imaging and outcomes following aneurysmal subarachnoid haemorrhage (aSAH). CONCLUSION Radiological biomarkers offer a potential non-invasive prognostication tool and may also allow early identifications of patients who may be at risk of DCI.
Collapse
|
12
|
Strickland BA, Bakhsheshian J, Emmanuel B, Amar A, Giannotta SL, Russin JJ, Mack W. Neuroprotective effect of minocycline against acute brain injury in clinical practice: A systematic review. J Clin Neurosci 2021; 86:50-57. [PMID: 33775346 DOI: 10.1016/j.jocn.2021.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/03/2020] [Accepted: 01/07/2021] [Indexed: 01/19/2023]
Abstract
Acute brain injury is a leading cause of morbidity and mortality worldwide. The term is inclusive of traumatic brain injury, cerebral ischemia, subarachnoid hemorrhage, and intracerebral hemorrhage. Current pharmacologic treatments have had minimal effect on improving neurological outcomes leading to a significant interest in the development neuroprotective agents. Minocycline is a second-generation tetracycline with high blood brain barrier penetrance due to its lipophilic properties. It functions across multiple molecular pathways involved in secondary-injury cascades following acute brain injury. Animal model studies suggest that minocycline might lead to improved neurologic outcomes, but few such trials exist in humans. Clinical investigations have been limited to small randomized trials in ischemic stroke patients which have not demonstrated a clear advantage in neurologic outcomes, but also have not been sufficiently powered to draw definitive conclusions. The potential neuroprotective effect of minocycline in the setting of traumatic brain injury, subarachnoid hemorrhage, and intracerebral hemorrhage have all been limited to pilot studies with phase II/III investigations pending. The authors aim to synthesize what is currently known about minocycline as a neuroprotective agent against acute brain injury in humans.
Collapse
Affiliation(s)
- Ben A Strickland
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA.
| | - Joshua Bakhsheshian
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA
| | - Ben Emmanuel
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA
| | - Arun Amar
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA
| | - Steven L Giannotta
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA
| | - Jonathan J Russin
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA
| | - William Mack
- Department of Neurosurgery, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
13
|
Hanmin C, Xiangyue Z, Lenahan C, Ling W, Yibo O, Yue H. Pleiotropic Role of Tenascin-C in Central Nervous System Diseases: From Basic to Clinical Applications. Front Neurol 2020; 11:576230. [PMID: 33281711 PMCID: PMC7691598 DOI: 10.3389/fneur.2020.576230] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
The extracellular matrix is composed of a variety of macromolecular substances secreted by cells, which form a complex network that supports and connects tissue structures, regulates the morphogenesis of tissues, and maintains the physiological activities of cells. Tenascin-C, a secreted extracellular matrix glycoprotein, is abundantly expressed after exposure to pathological stimuli. It plays an important regulatory role in brain tumors, vascular diseases, and neurodegenerative diseases by mediating inflammatory responses, inducing brain damage, and promoting cell proliferation, migration, and angiogenesis through multiple signaling pathways. Therefore, tenascin-C may become a potential therapeutic target for intracranial diseases. Here, we review and discuss the latest literature regarding tenascin-C, and we comprehensively explain the role and clinical significance of tenascin-C in intracranial diseases.
Collapse
Affiliation(s)
- Chen Hanmin
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Xiangyue
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Wang Ling
- Department of Operating Room, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ou Yibo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - He Yue
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Hasegawa Y, Cheng C, Hayashi K, Takemoto Y, Kim-Mitsuyama S. Anti-apoptotic effects of BDNF-TrkB signaling in the treatment of hemorrhagic stroke. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2020.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
15
|
Oka F, Chung DY, Suzuki M, Ayata C. Delayed Cerebral Ischemia After Subarachnoid Hemorrhage: Experimental-Clinical Disconnect and the Unmet Need. Neurocrit Care 2020; 32:238-251. [PMID: 30671784 PMCID: PMC7387950 DOI: 10.1007/s12028-018-0650-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Delayed cerebral ischemia (DCI) is among the most dreaded complications following aneurysmal subarachnoid hemorrhage (SAH). Despite advances in neurocritical care, DCI remains a significant cause of morbidity and mortality, prolonged intensive care unit and hospital stay, and high healthcare costs. Large artery vasospasm has classically been thought to lead to DCI. However, recent failure of clinical trials targeting vasospasm to improve outcomes has underscored the disconnect between large artery vasospasm and DCI. Therefore, interest has shifted onto other potential mechanisms such as microvascular dysfunction and spreading depolarizations. Animal models can be instrumental in dissecting pathophysiology, but clinical relevance can be difficult to establish. METHODS Here, we performed a systematic review of the literature on animal models of SAH, focusing specifically on DCI and neurological deficits. RESULTS We find that dog, rabbit and rodent models do not consistently lead to DCI, although some degree of delayed vascular dysfunction is common. Primate models reliably recapitulate delayed neurological deficits and ischemic brain injury; however, ethical issues and cost limit their translational utility. CONCLUSIONS To facilitate translation, clinically relevant animal models that reproduce the pathophysiology and cardinal features of DCI after SAH are urgently needed.
Collapse
Affiliation(s)
- Fumiaki Oka
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
- Department of Neurosurgery, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - David Y Chung
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Michiyasu Suzuki
- Department of Neurosurgery, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Cenk Ayata
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
16
|
Wu MY, Gao F, Yang XM, Qin X, Chen GZ, Li D, Dang BQ, Chen G. Matrix metalloproteinase-9 regulates the blood brain barrier via the hedgehog pathway in a rat model of traumatic brain injury. Brain Res 2020; 1727:146553. [DOI: 10.1016/j.brainres.2019.146553] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/31/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
|
17
|
Geraghty JR, Davis JL, Testai FD. Neuroinflammation and Microvascular Dysfunction After Experimental Subarachnoid Hemorrhage: Emerging Components of Early Brain Injury Related to Outcome. Neurocrit Care 2019; 31:373-389. [PMID: 31012056 PMCID: PMC6759381 DOI: 10.1007/s12028-019-00710-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aneurysmal subarachnoid hemorrhage has a high mortality rate and, for those who survive this devastating injury, can lead to lifelong impairment. Clinical trials have demonstrated that cerebral vasospasm of larger extraparenchymal vessels is not the sole contributor to neurological outcome. Recently, the focus of intense investigation has turned to mechanisms of early brain injury that may play a larger role in outcome, including neuroinflammation and microvascular dysfunction. Extravasated blood after aneurysm rupture results in a robust inflammatory response characterized by activation of microglia, upregulation of cellular adhesion molecules, recruitment of peripheral immune cells, as well as impaired neurovascular coupling, disruption of the blood-brain barrier, and imbalances in endogenous vasodilators and vasoconstrictors. Each of these phenomena is either directly or indirectly associated with neuronal death and brain injury. Here, we review recent studies investigating these various mechanisms in experimental models of subarachnoid hemorrhage with special emphasis on neuroinflammation and its effect on microvascular dysfunction. We discuss the various therapeutic targets that have risen from these mechanistic studies and suggest the utility of a multi-targeted approach to preventing delayed injury and improving outcome after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Joseph R Geraghty
- Department of Neurology and Rehabilitation, College of Medicine, University of Illinois at Chicago, 912 S. Wood St. Suite 174N, Chicago, IL, 60612, USA.
- Medical Scientist Training Program, University of Illinois at Chicago, Chicago, IL, USA.
| | - Joseph L Davis
- Department of Neurology and Rehabilitation, College of Medicine, University of Illinois at Chicago, 912 S. Wood St. Suite 174N, Chicago, IL, 60612, USA
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, College of Medicine, University of Illinois at Chicago, 912 S. Wood St. Suite 174N, Chicago, IL, 60612, USA
| |
Collapse
|
18
|
Noël G, Tham DKL, MacVicar BA, Moukhles H. Agrin plays a major role in the coalescence of the aquaporin-4 clusters induced by gamma-1-containing laminin. J Comp Neurol 2019; 528:407-418. [PMID: 31454080 DOI: 10.1002/cne.24763] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/13/2019] [Accepted: 08/22/2019] [Indexed: 01/06/2023]
Abstract
The basement membrane that seperates the endothelial cells and astrocytic endfeet that comprise the blood-brain barrier is rich in collagen, laminin, agrin, and perlecan. Previous studies have demonstrated that the proper recruitment of the water-permeable channel aquaporin-4 (AQP4) to astrocytic endfeet is dependent on interactions between laminin and the receptor dystroglycan. In this study, we conducted a deeper investigation into how the basement membrane might further regulate the expression, localization, and function of AQP4, using primary astrocytes as a model system. We found that treating these cells with laminin causes endogenous agrin to localize to the cell surface, where it co-clusters with β-dystroglycan (β-DG). Conversely, agrin sliencing profoundly disrupts β-DG clustering. As in the case of laminin111, Matrigel™, a complete basement membrane analog, also causes the clustering of AQP4 and β-DG. This clustering, whether induced by laminin111 or Matrigel™ is inhibited when the astrocytes are first incubated with an antibody against the γ1 subunit of laminin, suggesting that the latter is crucial to the process. Finally, we showed that laminin111 appears to negatively regulate AQP4-mediated water transport in astrocytes, suppressing the cell swelling that occurs following a hypoosmotic challenge. This suppression is abolished if DG expression is silenced, again demonstrating the central role of this receptor in relaying the effects of laminin.
Collapse
Affiliation(s)
- Geoffroy Noël
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel Kai Long Tham
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian A MacVicar
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hakima Moukhles
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
Liu L, Zhang P, Zhang Z, Hu Q, He J, Liu H, Zhao J, Liang Y, He Z, Li X, Sun X, Guo Z. LXA4 ameliorates cerebrovascular endothelial dysfunction by reducing acute inflammation after subarachnoid hemorrhage in rats. Neuroscience 2019; 408:105-114. [DOI: 10.1016/j.neuroscience.2019.03.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/15/2019] [Accepted: 03/15/2019] [Indexed: 12/18/2022]
|
20
|
Vadokas G, Koehler S, Weiland J, Lilla N, Stetter C, Westermaier T. Early Antiinflammatory Therapy Attenuates Brain Damage After Sah in Rats. Transl Neurosci 2019; 10:104-111. [PMID: 31098320 PMCID: PMC6487785 DOI: 10.1515/tnsci-2019-0018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/18/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Early inflammatory processes may play an important role in the development of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Experimental studies suggest that anti-inflammatory and membrane-stabilizing drugs might have beneficial effects, although the underlying mechanisms are not fully understood. The aim of this study was to investigate the effect of early treatment with methylprednisolone and minocycline on cerebral perfusion and EBI after experimental SAH. METHODS Male Sprague-Dawley rats were subjected to SAH using the endovascular filament model. 30 minutes after SAH, they were randomly assigned to receive an intravenous injection of methylprednisolone (16mg/kg body weight, n=10), minocycline (45mg/kg body weight, n=10) or saline (n=11). Mean arterial blood pressure (MABP), intracranial pressure (ICP) and local cerebral blood flow (LCBF) over both hemispheres were recorded continuously for three hours following SAH. Neurological assessment was performed after 24 hours. Hippocampal damage was analyzed by immunohistochemical staining (caspase 3). RESULTS Treatment with methylprednisolone or minocycline did not result in a significant improvement of MABP, ICP or LCBF. Animals of both treatment groups showed a non-significant trend to better neurological recovery compared to animals of the control group. Mortality was reduced and hippocampal damage significantly attenuated in both methylprednisolone and minocycline treated animals. CONCLUSION The results of this study suggest that inflammatory processes may play an important role in the pathophysiology of EBI after SAH. Early treatment with the anti-inflammatory drugs methylprednisolone or minocycline in the acute phase of SAH has the potential to reduce brain damage and exert a neuroprotective effect.
Collapse
Affiliation(s)
- Georg Vadokas
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
- Department of Urology, Canisius Wilhelmina Hospital Nijmegen, Weg door Jonkerbos 100, 6532 SZ Nijmegen, Netherlands
| | - Stefan Koehler
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Judith Weiland
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Nadine Lilla
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Christian Stetter
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| | - Thomas Westermaier
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080Würzburg, Germany
| |
Collapse
|
21
|
Chen D, Huang C, Chen Z. A review for the pharmacological effect of lycopene in central nervous system disorders. Biomed Pharmacother 2019; 111:791-801. [DOI: 10.1016/j.biopha.2018.12.151] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/31/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
|
22
|
Tortuosity of the Internal Carotid Artery and Its Clinical Significance in the Development of Aneurysms. J Clin Med 2019; 8:jcm8020237. [PMID: 30759737 PMCID: PMC6406528 DOI: 10.3390/jcm8020237] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/16/2019] [Accepted: 02/01/2019] [Indexed: 02/03/2023] Open
Abstract
Tortuosity of blood vessels is a common angiographic finding that may indicate systemic disease and can be correlated with vascular pathologies. In this work, we determined whether patients with and without internal carotid artery (ICA) aneurysm presented with differences in its tortuosity descriptors. We retrospectively analysed data of 298 patients hospitalized between January 2014 and June 2018. For each patient's imaging data, we extracted a curve representing the ICA course and measured its Relative Length (RL), Sum of Angle Metrics (SOAM), Product of Angle Distance (PAD), Triangular Index (TI), and Inflection Count Metrics (ICM). We found that patients with an ICA aneurysm had significantly lower RL (0.46 ± 0.19 vs. 0.51 ± 0.17; p = 0.023) and significantly higher SOAM (0.39 ± 0.21 vs. 0.32 ± 0.21 p = 0.003), PAD (0.38 ± 0.19 vs. 0.32 ± 0.21; p = 0.011), TI (0.30 ± 0.11 vs. 0.27 ± 0.14; p = 0.034), and ICM (0.30 ± 0.16 vs. 0.22 ± 0.12; p < 0.001). We found that that patients who presented with a subarachnoid hemorrhage had significantly higher PAD (0.46 ± 0.22 vs. 0.35 ± 0.20; p = 0.024). In conclusion, higher tortuosity of ICA is associated with ICA aneurysm presence.
Collapse
|
23
|
Li T, Xu W, Gao L, Guan G, Zhang Z, He P, Xu H, Fan L, Yan F, Chen G. Mesencephalic astrocyte-derived neurotrophic factor affords neuroprotection to early brain injury induced by subarachnoid hemorrhage via activating Akt-dependent prosurvival pathway and defending blood-brain barrier integrity. FASEB J 2018; 33:1727-1741. [PMID: 30211660 DOI: 10.1096/fj.201800227rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
This study aimed to explore the neuroprotective effect of mesencephalic astrocyte-derived neurotrophic factor (MANF) protein on early brain injury caused by subarachnoid hemorrhage (SAH) and the relevant mechanisms in experimental rats, expecting to understand whether MANF was a potential therapeutic target for SAH treatment. A perforation model of SAH was introduced into the study. Recombinant human MANF (rh-MANF) and protein kinase B (Akt) inhibitor (MK2206) were used to explore the effect and the mechanisms. Multiple approaches for systemic assessment were employed in the research, including the Garcia test, the SAH grade, Evans blue (EB) dye leakage, brain-water content (BWC), the rotarod test, and the Morris water-navigation task, as were biotechniques, such as immunohistochemistry, Western blot, transmission electron microscopy, and flow cytometry. MANF was mainly expressed in rat neurons, and its expression increased significantly at 3 h after SAH induction and peaked at 24 h. Stereotactic injection of rh-MANF into the cerebroventricle significantly increased the level of MANF, p-Akt, p-mouse double minute 2 homolog (p-MDM2), and B-cell lymphoma 2 (Bcl-2) in brain tissue, whereas it down-regulated the expression of P53, Bcl-2-associated X protein (Bax), and cleaved caspase-3, which indicated that neuronal apoptosis was remarkably suppressed. Expression of matrix metallopeptidase 9 (MMP-9) was also suppressed by the rh-MANF injection. Furthermore, neurologic deficits, EB dye leakage, and BWC were reduced, and long-lasting neuroprotection was noted with rh-MANF administration. The antiapoptotic and blood-brain barrier (BBB) protective effect could be offset by administering MK2206. MANF could alleviate neuronal apoptosis by activating Akt-dependent prosurvival pathway and abate BBB damage via MMP-9 suppression. MANF showed not only transient but also long-lasting neuroprotective properties. The rh-MANF as a potential drug for treating SAH might be of clinical use.-Li, T., Xu, W., Gao, L., Guan, G., Zhang, Z., He, P., Xu, H., Fan, L., Yan, F., Chen, G. Mesencephalic astrocyte-derived neurotrophic factor affords neuroprotection to early brain injury induced by subarachnoid hemorrhage via activating Akt-dependent prosurvival pathway and defending blood-brain barrier integrity.
Collapse
Affiliation(s)
- Tao Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liansheng Gao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guoping Guan
- Department of Neurosurgery, Fenghua Hospital, Ningbo, China
| | - Zhongyuan Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Pingyou He
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hangzhe Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Linfeng Fan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Blackburn SL, Swisher CB, Grande AW, Rubi A, Verbick LZ, McCabe A, Lad SP. Novel Dual Lumen Catheter and Filtration Device for Removal of Subarachnoid hemorrhage: First Case Report. Oper Neurosurg (Hagerstown) 2018; 16:E148-E153. [DOI: 10.1093/ons/opy151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/16/2018] [Indexed: 11/13/2022] Open
Abstract
Abstract
BACKGROUND AND IMPORTANCE
The amount of subarachnoid blood and the presence of toxic blood breakdown products in the cerebrospinal fluid (CSF) have long been associated with poor outcomes in aneurysmal subarachnoid hemorrhage. The Neurapheresis™ system (Minnetronix Inc, St. Paul, Minnesota) has been developed to filter CSF and remove blood products, and is being investigated for safety and feasibility in the ExtracorPoreal FILtration of subarachnoid hemorrhage via SpinaL CAtheteR (PILLAR) study. We report the first case using this novel device.
CLINICAL PRESENTATION
A 65-yr-old female presented with a ruptured left posterior communicating artery aneurysm. Following placement of a ventriculostomy and coil embolization of her aneurysm, the patient underwent placement of a lumbar dual lumen catheter for CSF filtration as part of the PILLAR study. In this case, a total of 9 h of filtration during 31 h of catheter indwelling resulted in 309.47 mL of processed CSF without complication. Computed tomography images demonstrated an interval reduction of subarachnoid hemorrhage immediately after filtration. The patient was discharged home on postbleed day 11 and at 30 d showed good recovery.
CONCLUSION
Safety of the Neurapheresis procedure was confirmed in this first case, and we will continue to evaluate safety of the Neurapheresis system through the PILLAR trial.
Collapse
Affiliation(s)
- Spiros L Blackburn
- Department of Neurosurgery, University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Christa B Swisher
- Department of Neurology, Duke University Medical Center, Durham, North Carolina
| | - Andrew W Grande
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota
| | - Alba Rubi
- Department of Neurosurgery, University of Texas Health Sciences Center at Houston, Houston, Texas
| | | | | | - Shivanand P Lad
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
25
|
Blackburn SL, Kumar PT, McBride D, Zeineddine HA, Leclerc J, Choi HA, Dash PK, Grotta J, Aronowski J, Cardenas JC, Doré S. Unique Contribution of Haptoglobin and Haptoglobin Genotype in Aneurysmal Subarachnoid Hemorrhage. Front Physiol 2018; 9:592. [PMID: 29904350 PMCID: PMC5991135 DOI: 10.3389/fphys.2018.00592] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/02/2018] [Indexed: 01/12/2023] Open
Abstract
Survivors of cerebral aneurysm rupture are at risk for significant morbidity and neurological deficits. Much of this is related to the effects of blood in the subarachnoid space which induces an inflammatory cascade with numerous downstream consequences. Recent clinical trials have not been able to reduce the toxic effects of free hemoglobin or improve clinical outcome. One reason for this may be the inability to identify patients at high risk for neurologic decline. Recently, haptoglobin genotype has been identified as a pertinent factor in diabetes, sickle cell, and cardiovascular disease, with the Hp 2-2 genotype contributing to increased complications. Haptoglobin is a protein synthesized by the liver that binds free hemoglobin following red blood cell lysis, and in doing so, prevents hemoglobin induced toxicity and facilitates clearance. Clinical studies in patients with subarachnoid hemorrhage indicate that Hp 2-2 patients may be a high-risk group for hemorrhage related complications and poor outcome. We review the relevance of haptoglobin in subarachnoid hemorrhage and discuss the effects of genotype and expression levels on the known mechanisms of early brain injury (EBI) and cerebral ischemia after aneurysm rupture. A better understanding of haptoglobin and its role in preventing hemoglobin related toxicity should lead to novel therapeutic avenues.
Collapse
Affiliation(s)
- Spiros L Blackburn
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Peeyush T Kumar
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Devin McBride
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Hussein A Zeineddine
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Jenna Leclerc
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States
| | - H Alex Choi
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - Pramod K Dash
- Department of Neurosurgery, The University of Texas Houston Health Sciences Center, Houston, TX, United States
| | - James Grotta
- Department of Neurology, The University of Texas Health Sciences Center, Houston, TX, United States
| | - Jaroslaw Aronowski
- Department of Neurology, The University of Texas Health Sciences Center, Houston, TX, United States
| | - Jessica C Cardenas
- Department of Surgery, Division of Acute Care Surgery and Center for Translational Injury Research, The University of Texas Health Science Center, Houston, TX, United States
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, College of Medicine, Gainesville, FL, United States.,Departments of Neurology, Psychiatry, Psychology, Pharmaceutics, and Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, FL, United States
| |
Collapse
|
26
|
Early matrix metalloproteinase-9 concentration in the first 48 h after aneurysmal subarachnoid haemorrhage predicts delayed cerebral ischaemia: An observational study. Eur J Anaesthesiol 2018; 33:662-9. [PMID: 27355865 DOI: 10.1097/eja.0000000000000494] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Delayed cerebral ischaemia from vasospasm is an important cause of complications and death after aneurysmal subarachnoid haemorrhage. There is currently no established biomarker for identifying patients at high risk of delayed cerebral ischaemia. OBJECTIVE Considering the important role of inflammation in the pathogenesis of delayed cerebral ischaemia, we investigated whether matrix metalloproteinase-9 (MMP-9) may be an efficient biomarker for predicting elayed cerebral ischaemia after subarachnoid haemorrhage. DESIGN Single-centre prospective observational study. SETTING Neuroscience Critical Care Unit of a teaching hospital. PARTICIPANTS Thirty consecutive patients with severe subarachnoid haemorrhage requiring external ventricular drainage were enrolled during 2013 and 2014. INTERVENTIONS Blood and cerebrospinal fluid (CSF) were sampled within the first 24 h and between 48 and 72 h after admission. We evaluated the activity and concentrations of MMP-9 and endothelin-1 with zymography and ELISA. Patients were allocated to groups with delayed cerebral ischaemia (n = 16) or without delayed cerebral ischaemia (n = 14). RESULTS Within 24 h, median [interquartile range] MMP-9 concentrations in CSF were significantly higher in patients with delayed cerebral ischaemia (47 [21 to 102] ng ml) than in those without delayed cerebral ischaemia (4 [2 to 13] ng ml, P = 0.001). CSF MMP-9 activity and endothelin-1 concentrations were correlated (r = 0.6, P = 0.02). The areas under the receiver operating characteristic curves were 0.73 (95% confidence interval [0.53 to 0.87]) and 0.91 (95% confidence interval [0.75 to 0.98]) for MMP-9 concentrations in plasma and CSF, respectively, at 24 h to predict delayed cerebral ischaemia CSF MMP-9 concentrations more than 14.3 ng ml at 24 h predicted the occurrence of delayed cerebral ischaemia with a sensitivity and specificity of 88 and 86%, respectively. After multivariate logistic analysis, only CSF MMP-9 concentrations at 24 h predicted the occurrence of delayed cerebral ischaemia (P = 0.01). CONCLUSION MMP-9 concentrations in both plasma and CSF, measured within 48 h after subarachnoid haemorrhage, were highly predictive of the occurrence of delayed cerebral ischaemia within the first 2 weeks. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT02397759.
Collapse
|
27
|
Liu L, Suzuki H. The Role of Matricellular Proteins in Experimental Subarachnoid Hemorrhage-Induced Early Brain Injury. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-3-319-66679-2_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
|
28
|
Frontera JA, Provencio JJ, Sehba FA, McIntyre TM, Nowacki AS, Gordon E, Weimer JM, Aledort L. The Role of Platelet Activation and Inflammation in Early Brain Injury Following Subarachnoid Hemorrhage. Neurocrit Care 2017; 26:48-57. [PMID: 27430874 DOI: 10.1007/s12028-016-0292-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Early brain injury (EBI) following aneurysmal subarachnoid hemorrhage (SAH) is an important predictor of poor functional outcome, yet the underlying mechanism is not well understood. Animal studies suggest that platelet activation and inflammation with subsequent microthrombosis and ischemia may be a mechanism of EBI. METHODS A prospective, hypothesis-driven study of spontaneous, SAH patients and controls was conducted. Platelet activation [thromboelastography maximum amplitude (MA)] and inflammation [C-reactive protein (CRP)] were measured serially over time during the first 72 h following SAH onset. Platelet activation and inflammatory markers were compared between controls and SAH patients with mild [Hunt-Hess (HH) 1-3] versus severe (HH 4-5) EBI. The association of these biomarkers with 3-month functional outcomes was evaluated. RESULTS We enrolled 127 patients (106 SAH; 21 controls). Platelet activation and CRP increased incrementally with worse EBI/HH grade, and both increased over 72 h (all P < 0.01). Both were higher in severe versus mild EBI (MA 68.9 vs. 64.8 mm, P = 0.001; CRP 12.5 vs. 1.5 mg/L, P = 0.003) and compared to controls (both P < 0.003). Patients with delayed cerebral ischemia (DCI) had more platelet activation (66.6 vs. 64.9 in those without DCI, P = 0.02) within 72 h of ictus. At 3 months, death or severe disability was more likely with higher levels of platelet activation (mRS4-6 OR 1.18, 95 % CI 1.05-1.32, P = 0.007) and CRP (mRS4-6 OR 1.02, 95 % CI 1.00-1.03, P = 0.041). CONCLUSIONS Platelet activation and inflammation occur acutely after SAH and are associated with worse EBI, DCI and poor 3-month functional outcomes. These markers may provide insight into the mechanism of EBI following SAH.
Collapse
Affiliation(s)
- Jennifer A Frontera
- Cerebrovascular Center of the Neurological Institute, Cleveland Clinic, 9500 Euclid Ave. S80, Cleveland, OH, 44195, USA. .,Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - J Javier Provencio
- Department of Neurology and Neuroscience, Brain Immunology and Glia Center, University of Virginia, Charlottesville, VA, USA
| | - Fatima A Sehba
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY, USA
| | - Thomas M McIntyre
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Amy S Nowacki
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Errol Gordon
- Department of Critical Care, Mount Sinai School of Medicine, New York, NY, USA
| | - Jonathan M Weimer
- Cerebrovascular Center of the Neurological Institute, Cleveland Clinic, 9500 Euclid Ave. S80, Cleveland, OH, 44195, USA
| | - Louis Aledort
- Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
29
|
van der Kleij LA, De Vis JB, Olivot JM, Calviere L, Cognard C, Zuithoff NPA, Rinkel GJE, Hendrikse J, Vergouwen MDI. Magnetic Resonance Imaging and Cerebral Ischemia After Aneurysmal Subarachnoid Hemorrhage. Stroke 2017; 48:239-245. [DOI: 10.1161/strokeaha.116.011707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 04/30/2016] [Accepted: 11/15/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Lisa A van der Kleij
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Jill B De Vis
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Jean-Marc Olivot
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Lionel Calviere
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Christophe Cognard
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Nicolaas P A Zuithoff
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Gabriel J E Rinkel
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Jeroen Hendrikse
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France
| | - Mervyn D I Vergouwen
- From the Department of Radiology (L.A.v.d.K., J.B.D.V., J.H.), Julius Center for Health Sciences and Primary Care (N.P.A.Z.), and Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus (G.J.E.R., M.D.I.V.), University Medical Center Utrecht, The Netherlands; and Department of Neurology (J.M.O., L.C.) and Department of Radiology (C.C.), University of Toulouse, France.
| |
Collapse
|
30
|
Burrell C, Avalon NE, Siegel J, Pizzi M, Dutta T, Charlesworth MC, Freeman WD. Precision medicine of aneurysmal subarachnoid hemorrhage, vasospasm and delayed cerebral ischemia. Expert Rev Neurother 2016; 16:1251-1262. [PMID: 27314601 DOI: 10.1080/14737175.2016.1203257] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Precision medicine provides individualized treatment of diseases through leveraging patient-to-patient variation. Aneurysmal subarachnoid hemorrhage carries tremendous morbidity and mortality with cerebral vasospasm and delayed cerebral ischemia proving devastating and unpredictable. Lack of treatment measures for these conditions could be improved through precision medicine. Areas covered: Discussed are the pathophysiology of CV and DCI, treatment guidelines, and evidence for precision medicine used for prediction and prevention of poor outcomes following aSAH. A PubMed search was performed using keywords cerebral vasospasm or delayed cerebral ischemia and either biomarkers, precision medicine, metabolomics, proteomics, or genomics. Over 200 peer-reviewed articles were evaluated. The studies presented cover biomarkers identified as predictive markers or therapeutic targets following aSAH. Expert commentary: The biomarkers reviewed here correlate with CV, DCI, and neurologic outcomes after aSAH. Though practical use in clinical management of aSAH is not well established, using these biomarkers as predictive tools or therapeutic targets demonstrates the potential of precision medicine.
Collapse
Affiliation(s)
| | - Nicole E Avalon
- a Department of Neurology , Mayo Clinic , Jacksonville , FL , USA
| | - Jason Siegel
- a Department of Neurology , Mayo Clinic , Jacksonville , FL , USA
| | - Michael Pizzi
- a Department of Neurology , Mayo Clinic , Jacksonville , FL , USA
| | - Tumpa Dutta
- b Endocrine Research Unit , Mayo Clinic , Rochester , MN , USA
| | | | | |
Collapse
|
31
|
Ethyl pyruvate alleviates early brain injury following subarachnoid hemorrhage in rats. Acta Neurochir (Wien) 2016; 158:1069-76. [PMID: 27072679 DOI: 10.1007/s00701-016-2795-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/27/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Previous studies have demonstrated the neuroprotective effects of ethyl pyruvate in central nervous system (CNS) diseases. However, whether ethyl pyruvate attenuates early brain injury after subarachnoid hemorrhage (SAH) remains unknown. This study was conducted to investigate the potential effects of ethyl pyruvate on early brain injury induced by SAH and explore the underlying mechanisms. METHODS Eighty-eight male Sprague-Dawley rats were used. An SAH model was induced by endovascular perforation. Ethyl pyruvate (100 mg/kg) or a vehicle was administered intraperitoneally at 1 h after SAH induction. SAH grade, neurological scores, brain water content, Evans blue extravasation, Western blots, and immunofluorescence were used to study the mechanisms of ethyl pyruvate. RESULTS Ethyl pyruvate treatment inhibited microglia activation and reduced the expression of proinflammatory cytokines (IL-1β and TNF-α). Ethyl pyruvate treatment also prevented disruption of tight junction proteins (occluding and claudin-5) and reduced expression of MMP-9. In addition, ethyl pyruvate treatment markedly reduced TUNEL-positive cells and expression of cleaved caspase-3. CONCLUSIONS Our results indicated that ethyl pyruvate treatment attenuated early brain injury and improved neurological function after SAH by inhibiting microglia activation and apoptosis and stabilizing the BBB.
Collapse
|
32
|
Glial cell response after aneurysmal subarachnoid hemorrhage — Functional consequences and clinical implications. Biochim Biophys Acta Mol Basis Dis 2016; 1862:492-505. [DOI: 10.1016/j.bbadis.2015.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 12/17/2022]
|
33
|
Fujimoto M, Shiba M, Kawakita F, Liu L, Shimojo N, Imanaka-Yoshida K, Yoshida T, Suzuki H. Deficiency of tenascin-C and attenuation of blood-brain barrier disruption following experimental subarachnoid hemorrhage in mice. J Neurosurg 2015; 124:1693-702. [PMID: 26473781 DOI: 10.3171/2015.4.jns15484] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECT Tenascin-C (TNC), a matricellular protein, is induced in the brain following subarachnoid hemorrhage (SAH). The authors investigated if TNC causes brain edema and blood-brain barrier (BBB) disruption following experimental SAH. METHODS C57BL/6 wild-type (WT) or TNC knockout (TNKO) mice were subjected to SAH by endovascular puncture. Ninety-seven mice were randomly allocated to WT sham-operated (n = 16), TNKO sham-operated (n = 16), WT SAH (n = 34), and TNKO SAH (n = 31) groups. Mice were examined by means of neuroscore and brain water content 24-48 hours post-SAH; and Evans blue dye extravasation and Western blotting of TNC, matrix metalloproteinase (MMP)-9, and zona occludens (ZO)-1 at 24 hours post-SAH. As a separate study, 16 mice were randomized to WT sham-operated, TNKO sham-operated, WT SAH, and TNKO SAH groups (n = 4 in each group), and activation of mitogen-activated protein kinases (MAPKs) was immunohistochemically evaluated at 24 hours post-SAH. Moreover, 40 TNKO mice randomly received an intracerebroventricular injection of TNC or phosphate-buffered saline, and effects of exogenous TNC on brain edema and BBB disruption following SAH were studied. RESULTS Deficiency of endogenous TNC prevented neurological impairments, brain edema formation, and BBB disruption following SAH; it was also associated with the inhibition of both MMP-9 induction and ZO-1 degradation. Endogenous TNC deficiency also inhibited post-SAH MAPK activation in brain capillary endothelial cells. Exogenous TNC treatment abolished the neuroprotective effects shown in TNKO mice with SAH. CONCLUSIONS Tenascin-C may be an important mediator in the development of brain edema and BBB disruption following SAH, mechanisms for which may involve MAPK-mediated MMP-9 induction and ZO-1 degradation. TNC could be a molecular target against which to develop new therapies for SAH-induced brain injuries.
Collapse
Affiliation(s)
| | | | | | - Lei Liu
- Departments of 1 Neurosurgery and
| | - Naoshi Shimojo
- Pathology and Matrix Biology.,Research Center for Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kyoko Imanaka-Yoshida
- Pathology and Matrix Biology.,Research Center for Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshimichi Yoshida
- Pathology and Matrix Biology.,Research Center for Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidenori Suzuki
- Departments of 1 Neurosurgery and.,Research Center for Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
34
|
Egashira Y, Zhao H, Hua Y, Keep RF, Xi G. White Matter Injury After Subarachnoid Hemorrhage: Role of Blood-Brain Barrier Disruption and Matrix Metalloproteinase-9. Stroke 2015; 46:2909-15. [PMID: 26374478 DOI: 10.1161/strokeaha.115.010351] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/31/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE We recently observed early white matter injury after experimental subarachnoid hemorrhage (SAH), but the underlying mechanisms are uncertain. This study investigated the potential role of matrix metalloproteinase (MMP)-9 in blood-brain barrier (BBB) disruption and consequent white matter injury. METHODS SAH was induced by endovascular perforation in adult male mice. The following 3 experiments were devised: (1) mice underwent magnetic resonance imaging at 24 h after SAH and were euthanized to determine BBB disruption and MMP-9 activation in white matter; (2) to investigate the role of MMP-9 in BBB disruption, lesion volumes on magnetic resonance imaging were compared between wild-type (WT) and MMP-9 knockout (MMP-9-/-) mice at 24 h after SAH; (3) WT and MMP-9-/- mice underwent magnetic resonance imaging at 1 and 8 days after SAH to detect time-dependent changes in brain injury. Brains were used to investigate myelin integrity in white matter. RESULTS In WT mice with SAH, white matter showed BBB disruption (albumin leakage) and T2 hyperintensity on magnetic resonance imaging. MMP-9 activity was elevated at 24 h after SAH. MMP-9-/- mice had less white matter T2 hyperintensity after SAH than WT mice. At 8 days after SAH, WT mice had decreased myelin integrity and MMP-9-/- mice developed less white matter injury. CONCLUSIONS SAH causes BBB disruption and consequent injury in white matter. MMP-9 plays an important role in those pathologies and could be a therapeutic target for SAH-induced white matter injury.
Collapse
Affiliation(s)
- Yusuke Egashira
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Hao Zhao
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Ya Hua
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Richard F Keep
- From the Department of Neurosurgery, University of Michigan, Ann Arbor
| | - Guohua Xi
- From the Department of Neurosurgery, University of Michigan, Ann Arbor.
| |
Collapse
|
35
|
Hasegawa Y, Suzuki H, Uekawa K, Kawano T, Kim-Mitsuyama S. Characteristics of Cerebrovascular Injury in the Hyperacute Phase After Induced Severe Subarachnoid Hemorrhage. Transl Stroke Res 2015; 6:458-66. [DOI: 10.1007/s12975-015-0423-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/30/2015] [Accepted: 09/01/2015] [Indexed: 10/23/2022]
|
36
|
Abstract
Early brain injury (EBI) plays a crucial role in the pathological progress of subarachnoid hemorrhage (SAH). This study was designed to determine whether rosiglitazone protects the brain against EBI in rats, and discuss the role of the anti-apoptotic mechanism mediated by Bcl-2 family proteins in this neuroprotection. 86 male Sprague-Dawley rats were divided into the sham group, the SAH+ vehicle group and the SAH+ rosiglitazone group. SAH was induced via an endovascular perforation technique and rosiglitazone (3mg/kg) or vehicle was administered. Mortality, neurological scores, brain water content, Evans blue dye assay, TUNEL stain assay, Gelatin zymography, and western blot analysis were performed. Rosiglitazone significantly improved mortality, neurological scores, brain water content, blood brain barrier (BBB) and apoptosis compared with the vehicle group within 24h after SAH. The TUNEL staining assay demonstrated that apoptosis was ameliorated. Cleaved Caspase-3 and MMP-9 expression was reduced, whereas Bcl-2 and p-Bad was markedly preserved by rosiglitazone. A significant elevation of p-Akt was detected after rosiglitazone treatment. Our study demonstrated that rosiglitazone plays a neuroprotective role in EBI after SAH via attenuation of BBB disruption, brain edema and apoptosis.
Collapse
|
37
|
Li J, Chen J, Mo H, Chen J, Qian C, Yan F, Gu C, Hu Q, Wang L, Chen G. Minocycline Protects Against NLRP3 Inflammasome-Induced Inflammation and P53-Associated Apoptosis in Early Brain Injury After Subarachnoid Hemorrhage. Mol Neurobiol 2015; 53:2668-78. [DOI: 10.1007/s12035-015-9318-8] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/24/2015] [Indexed: 12/12/2022]
|
38
|
Kurogi R, Kikkawa Y, Matsuo S, Nakamizo A, Mizoguchi M, Sasaki T. Upregulation of tissue inhibitor of metalloproteinase-1 contributes to restoration of the extracellular matrix in the rabbit basilar artery during cerebral vasospasm after subarachnoid hemorrhage. Brain Res 2015; 1616:26-36. [PMID: 25940763 DOI: 10.1016/j.brainres.2015.04.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/06/2015] [Accepted: 04/23/2015] [Indexed: 11/17/2022]
Abstract
Vascular remodeling caused by extracellular matrix (ECM) metabolism contributes to the development of cerebral vasospasm after subarachnoid hemorrhage (SAH). The balance between tissue inhibitor of metalloproteinases (TIMPs) and matrix metalloproteinases (MMPs) plays an important role in ECM remodeling. We investigated the mechanism of vascular remodeling following cerebral vasospasm in a rabbit double hemorrhage model. Rabbit basilar arteries were harvested on days 3, 5, and 7 after initial hemorrhage. TIMP-1, TIMP-2, MMP-2, and MMP-9 mRNA and protein expression were investigated with microarray analysis, quantitative real-time PCR, immunoblot analysis, and enzyme-linked immunosorbent assay (ELISA). The expression and localization of TIMP-1, TIMP-2, MMP-2, MMP-9, elastin, fibronectin, laminin, and collagens I, III, and IV were investigated with immuohistochemical staining. After SAH, TIMP-1 mRNA and protein expression were significantly increased on day 3 and then decreased to the control level on days 5 and 7. MMP-9 protein expression was significantly increased on day 7. TIMP-2 and MMP-2 mRNA and protein expression were significantly increased on day 7. Elastin, fibronectin, laminin, and collagens I, III, and IV protein expression was decreased on day 3 and then restored to control levels on day 7. Upregulation of TIMP-1 during the early phase of cerebral vasospasm may contribute to the recovery of the ECM during the late phase of cerebral vasospasm, resulting in a protective role of TIMP-1 from cerebral vasospasm. Moreover, the increase in arterial compliance by the decrease in ECM during the early phase of cerebral vasospasm may facilitate vasoconstriction of the cerebral artery.
Collapse
Affiliation(s)
- Ryota Kurogi
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City 812-8582, Fukuoka, Japan
| | - Yuichiro Kikkawa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City 812-8582, Fukuoka, Japan; Department of Cerebrovascular Surgery, International Medical Center, Saitama Medical University, 1397-1 Yamane, Hidaka City 350-1298, Saitama, Japan.
| | - Satoshi Matsuo
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City 812-8582, Fukuoka, Japan
| | - Akira Nakamizo
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City 812-8582, Fukuoka, Japan; Department of Neurosurgery, Steel Memorial Yawata Hospital, 1-1-1 Harunomachi, Yahatahigashi-ku, Kitakyusyu City 805-8508, Fukuoka, Japan
| | - Masahiro Mizoguchi
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City 812-8582, Fukuoka, Japan
| | - Tomio Sasaki
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City 812-8582, Fukuoka, Japan
| |
Collapse
|
39
|
Protective actions of PJ34, a poly(ADP-ribose)polymerase inhibitor, on the blood-brain barrier after traumatic brain injury in mice. Neuroscience 2015; 291:26-36. [PMID: 25668593 DOI: 10.1016/j.neuroscience.2015.01.070] [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: 11/12/2014] [Revised: 01/30/2015] [Accepted: 01/30/2015] [Indexed: 01/12/2023]
Abstract
Poly(ADP-ribose) polymerase (PARP) is activated by oxidative stress and plays an important role in traumatic brain injury (TBI). The objective of this study was to investigate whether PARP activation participated in the blood-brain barrier (BBB) disruption and edema formation in a mouse model of controlled cortical impact (CCI). N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide (PJ34) (10 mg/kg), a selective PARP inhibitor, was administered intraperitoneally at 5 min and 8 h after experimental CCI. After 6 h and 24 h of CCI, the permeability of the cortical BBB was determined after Evans Blue administration. The water content of the brain was also measured. Treatment with PJ34 markedly attenuated the permeability of the BBB and decreased the brain edema at 6 h and 24 h after CCI. Our data showed the up-regulation of nuclear factor-κB in cytosolic fractions and nuclear fractions in the injured cortex, and these changes were reversed by PJ34. Moreover, PJ34 significantly lessened the activities of myeloperoxidase and the levels of matrix metalloproteinase-9, enhanced the levels of occludin, laminin, collagen IV and integrin β1, reduced neurological deficits, decreased the contusion volume, and attenuated the necrotic and apoptotic neuronal cell death. These data suggest the protective effects of PJ34 on BBB integrity and cell death during acute TBI.
Collapse
|
40
|
MMP-9 expression and activity is concurrent with endothelial cell apoptosis in the basilar artery after subarachnoid hemorrhaging in rats. Neurol Sci 2015; 36:1241-5. [DOI: 10.1007/s10072-015-2092-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/21/2015] [Indexed: 01/28/2023]
|
41
|
Pathological mechanisms underlying aneurysmal subarachnoid haemorrhage and vasospasmElsevier Ltd. J Clin Neurosci 2015; 22:1-5. [DOI: 10.1016/j.jocn.2014.05.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/27/2014] [Accepted: 05/04/2014] [Indexed: 11/18/2022]
|
42
|
Huenges Wajer IMC, Cremers CHP, van Zandvoort MJE, Vergouwen MDI, van der Schaaf IC, Velthuis BK, Dankbaar JW, Vos PC, Visser-Meily JMA, Rinkel GJE. CT perfusion on admission and cognitive functioning 3 months after aneurysmal subarachnoid haemorrhage. J Neurol 2014; 262:623-8. [DOI: 10.1007/s00415-014-7601-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/26/2014] [Accepted: 11/29/2014] [Indexed: 10/24/2022]
|
43
|
Chiu PS, Lai SC. Matrix metalloproteinase-9 leads to blood-brain barrier leakage in mice with eosinophilic meningoencephalitis caused by Angiostrongylus cantonensis. Acta Trop 2014; 140:141-50. [PMID: 25158284 DOI: 10.1016/j.actatropica.2014.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/04/2014] [Accepted: 08/16/2014] [Indexed: 11/29/2022]
Abstract
Blood-brain barrier (BBB) disruption is associated with tight junction protein degradation, basal membrane disruption, and astrocyte damage. This study aims to investigate the role of matrix metalloproteinase (MMP)-9 in BBB disruption during Angiostrongylus cantonensis infection. We used mice infected with A. cantonensis, in which parasite-induced eosinophilia and inflammation might induce MMP-9 elevation. MMP-9 could cause claudin-5 degradation in endothelium tight junction, collagen type IV degradation in basal membranes, and S100B degradation in astrocytes of wild-type mice. BBB permeability was significantly attenuated in MMP-9 knockout mice than in wild-type mice in angiostrongyliasis meningoencephalitis. Immune cell aggregates were also more attenuated in the brains of MMP-9 knockout mice than in the brains of wild-type mice. Results suggest that MMP-9 activities are significant in BBB disruption in angiostrongyliasis meningoencephalitis. This study improves understanding of molecular mechanisms that underlie brain invasion by A. cantonensis, which is a key step in the pathogenesis of meningoencephalitis, and can offer a new strategy to reduce mortality.
Collapse
Affiliation(s)
- Ping-Sung Chiu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Shih-Chan Lai
- Department of Parasitology, Chung Shan Medical University, Taichung 402, Taiwan; Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan.
| |
Collapse
|
44
|
Zhang T, Su J, Guo B, Zhu T, Wang K, Li X. Ursolic acid alleviates early brain injury after experimental subarachnoid hemorrhage by suppressing TLR4-mediated inflammatory pathway. Int Immunopharmacol 2014; 23:585-91. [PMID: 25466266 DOI: 10.1016/j.intimp.2014.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/08/2014] [Accepted: 10/08/2014] [Indexed: 10/24/2022]
Abstract
Our previous studies proved that ursolic acid (UA) protected against early brain injury (EBI) by modulating oxidative stress after experimental subarachnoid hemorrhage (SAH), but it has not been evaluated yet about its effects on an inflammatory pathway in a SAH model. This study was undertaken to evaluate the influence of UA on the toll-like receptor 4 (TLR4) signaling pathway after SAH. Adult male SD rats were divided into vehicle-treated sham, vehicle-treated SAH, and UA-treated SAH groups. The endovascular puncture model was used to induce SAH and all the rats were subsequently sacrificed at 48 h after SAH. UA administration markedly decreased the expressions of TLR4 pathway-related agents, such as intercellular adhesion molecule-1 (ICAM-1), TLR4, nuclear factor-κB (NF-κB) P65, interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS) and matrix metalloproteinase (MMP)-9. Apoptosis detected by TUNEL indicated that fewer positive cells appeared in UA administration SAH groups than the control group. In conclusion, UA may attenuate EBI after SAH in rats by suppressing the TLR4-mediated inflammatory pathway.
Collapse
|
45
|
Mattila OS, Rantanen V, Saksi J, Strbian D, Pikkarainen T, Hautaniemi S, Lindsberg PJ. Workflow for automated quantification of cerebromicrovascular gelatinase activity. Microvasc Res 2014; 97:19-24. [PMID: 25242681 DOI: 10.1016/j.mvr.2014.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/24/2014] [Accepted: 08/06/2014] [Indexed: 11/19/2022]
Abstract
The gelatinase enzymes, matrix metalloproteinases -2 and -9, are central mediators of blood-brain barrier disruption, actively studied in experimental models of neurological disease. Staining with in situ zymography (ISZ) allows visualization of gelatinase activity directly in brain tissue sections. However, quantifying microvascular gelatinase activity from ISZ-images is challenging and time consuming, as surrounding cell types often show significant confounding activity. We describe validation and performance of a workflow for automated image analysis of cerebromicrovascular gelatinase activity, now released for open-access use. In comparison to manual analysis, the automated workflow showed superior accuracy, was faster to execute and allows for more detailed analysis of heterogeneity in the microvasculature. We further suggest recommendations for quantifying and reporting this type of activity in experimental studies, focusing on ischemic stroke.
Collapse
Affiliation(s)
- Olli S Mattila
- Research Programs Unit, Molecular Neurology, and Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland.
| | - Ville Rantanen
- Research Programs Unit, Genome-Scale Biology, Institute of Biomedicine, Biochemistry and Developmental Biology, University of Helsinki, Finland
| | - Jani Saksi
- Research Programs Unit, Molecular Neurology, and Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Daniel Strbian
- Department of Neurology, Helsinki University Central Hospital, Finland
| | - Tero Pikkarainen
- Research Programs Unit, Molecular Neurology, and Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Sampsa Hautaniemi
- Research Programs Unit, Genome-Scale Biology, Institute of Biomedicine, Biochemistry and Developmental Biology, University of Helsinki, Finland
| | - Perttu J Lindsberg
- Research Programs Unit, Molecular Neurology, and Department of Clinical Neurosciences, University of Helsinki, Helsinki, Finland; Department of Neurology, Helsinki University Central Hospital, Finland
| |
Collapse
|
46
|
Rat endovascular perforation model. Transl Stroke Res 2014; 5:660-8. [PMID: 25213427 DOI: 10.1007/s12975-014-0368-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/18/2014] [Accepted: 08/21/2014] [Indexed: 12/31/2022]
Abstract
Experimental animal models of aneurysmal subarachnoid hemorrhage (SAH) have provided a wealth of information on the mechanisms of brain injury. The rat endovascular perforation (EVP) model replicates the early pathophysiology of SAH and hence is frequently used to study early brain injury following SAH. This paper presents a brief review of historical development of the EVP model and details the technique used to create SAH and considerations necessary to overcome technical challenges.
Collapse
|
47
|
Zhan Y, Krafft PR, Lekic T, Ma Q, Souvenir R, Zhang JH, Tang J. Imatinib preserves blood-brain barrier integrity following experimental subarachnoid hemorrhage in rats. J Neurosci Res 2014; 93:94-103. [PMID: 25196554 DOI: 10.1002/jnr.23475] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/22/2014] [Accepted: 07/24/2014] [Indexed: 01/30/2023]
Abstract
Blood-brain barrier (BBB) disruption and consequent edema formation contribute to the development of early brain injury following subarachnoid hemorrhage (SAH). Various cerebrovascular insults result in increased platelet-derived growth factor receptor (PDGFR)-α stimulation, which has been linked to BBB breakdown and edema formation. This study examines whether imatinib, a PDGFR inhibitor, can preserve BBB integrity in a rat endovascular perforation SAH model. Imatinib (40 or 120 mg/kg) or a vehicle was administered intraperitoneally at 1 hr after SAH induction. BBB leakage, brain edema, and neurological deficits were evaluated. Total and phosphorylated protein expressions of PDGFR-α, c-Src, c-Jun N-terminal kinase (JNK), and c-Jun were measured, and enzymatic activities of matrix metalloproteinase (MMP)-2 and MMP-9 were determined in the injured brain. Imatinib treatment significantly ameliorated BBB leakage and edema formation 24 hr after SAH, which was paralleled by improved neurological functions. Decreased brain expressions of phosphorylated PDGFR-α, c-Src, JNK, and c-Jun as well as reduced MMP-9 activities were found in treated animals. PDGFR-α inhibition preserved BBB integrity following experimental SAH; however, the protective mechanisms remain to be elucidated. Targeting PDGFR-α signaling might be advantageous to ameliorate early brain injury following SAH.
Collapse
Affiliation(s)
- Yan Zhan
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA.,Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Paul R Krafft
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Tim Lekic
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Qingyi Ma
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Rhonda Souvenir
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - John H Zhang
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Jiping Tang
- Department of Basic Sciences, Division of Physiology, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
48
|
Oudshoorn SC, Rinkel GJE, Molyneux AJ, Kerr RS, Dorhout Mees SM, Backes D, Algra A, Vergouwen MDI. Aneurysm treatment <24 versus 24-72 h after subarachnoid hemorrhage. Neurocrit Care 2014; 21:4-13. [PMID: 24639201 DOI: 10.1007/s12028-014-9969-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION In patients with aneurysmal subarachnoid hemorrhage (aSAH), it is unclear whether aneurysm treatment <24 h after ictus results in better outcomes than treatment 24-72 h after aSAH. We studied whether aneurysm occlusion <24 h is associated with better outcomes than occlusion 24-72 h after aSAH. METHODS We used two cohorts of patients with aSAH: (1) the UMC Utrecht cohort with patients admitted between 2008 and 2012 and (2) the International Subarachnoid Aneurysm Trial cohort. Aneurysm treatment was categorized into <24 h and 24-72 h after ictus. We calculated adjusted risk ratios (aRRs) with 95% confidence intervals (CIs) using Poisson regression analyses for poor functional outcome (death or dependency) for both cohorts separately, and performed a pooled analysis based on individual patient data. We also performed a worst-case scenario analysis wherein all patients with rebleeding >3 h after admission were re-categorized into the group with aneurysm treatment 24-72 h after aSAH. RESULTS We included 1,238 patients (UMC Utrecht cohort: n = 330; ISAT: n = 908). The aRR for poor outcome after treatment <24 h was in the UMC Utrecht cohort 1.84 (95% CI: 1.25-2.70), in ISAT 1.14 (95% CI 0.84-1.55), in the pooled analysis 1.37 (95% CI 1.11-1.68), and in the worst-case scenario pooled analysis 1.24 (95% CI 1.01-1.52). CONCLUSION Our results suggest that aneurysm occlusion can be performed in day time within 72 h after ictus, instead of on an emergency basis. However, due to the retrospective, non-randomized design of our study, our results cannot be considered as definitive evidence.
Collapse
Affiliation(s)
- Simone C Oudshoorn
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Room G03-228, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
49
|
To look beyond vasospasm in aneurysmal subarachnoid haemorrhage. BIOMED RESEARCH INTERNATIONAL 2014; 2014:628597. [PMID: 24967389 PMCID: PMC4055362 DOI: 10.1155/2014/628597] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/07/2014] [Indexed: 12/26/2022]
Abstract
Delayed cerebral vasospasm has classically been considered the most important and treatable cause of mortality and morbidity in patients with aneurysmal subarachnoid hemorrhage (aSAH). Secondary ischemia (or delayed ischemic neurological deficit, DIND) has been shown to be the leading determinant of poor clinical outcome in patients with aSAH surviving the early phase and cerebral vasospasm has been attributed to being primarily responsible. Recently, various clinical trials aimed at treating vasospasm have produced disappointing results. DIND seems to have a multifactorial etiology and vasospasm may simply represent one contributing factor and not the major determinant. Increasing evidence shows that a series of early secondary cerebral insults may occur following aneurysm rupture (the so-called early brain injury). This further aggravates the initial insult and actually determines the functional outcome. A better understanding of these mechanisms and their prevention in the very early phase is needed to improve the prognosis. The aim of this review is to summarize the existing literature on this topic and so to illustrate how the presence of cerebral vasospasm may not necessarily be a prerequisite for DIND development. The various factors determining DIND that worsen functional outcome and prognosis are then discussed.
Collapse
|
50
|
Plog BA, Moll KM, Kang H, Iliff JJ, Dashnaw ML, Nedergaard M, Vates GE. A novel technique for morphometric quantification of subarachnoid hemorrhage-induced microglia activation. J Neurosci Methods 2014; 229:44-52. [PMID: 24735531 DOI: 10.1016/j.jneumeth.2014.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 03/24/2014] [Accepted: 04/05/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a neurologic catastrophe and poor outcome is typically attributed to vasospasm; however, there is also evidence that SAH causes a pro-inflammatory state and these two phenomena may be interrelated. SAH causes activation of microglia, but the time course and degree of microglial activation after SAH and its link to poor patient outcome and vasospasm remains unknown. NEW METHOD Transgenic mice expressing eGFP under the control of the CX3CR1 locus, in which microglia are endogenously fluorescent, were randomly assigned to control or SAH groups. Immunohistochemistry for CD-68 and CD-31 was performed at different time points after SAH. Using confocal microscopy and MatLab software, we have developed a novel technique to detect and quantify the stages of microglial activation and return to quiescence using an automated computerized morphometric analysis. RESULTS We detected a statistically significant decrease in microglial process complexity 2 and 7 days following SAH. In addition, we detected a statistically significant increase in microglial domain volume 1 day following SAH; however, microglial domain volume returned to baseline by 2 days. COMPARISON WITH EXISTING METHOD Most techniques for microglia assessment are qualitative, not quantitative, and are therefore inadequate to address the effects of anti-inflammatory drug treatment or other therapies after SAH. CONCLUSIONS Using novel image analysis techniques we were able to reproducibly quantify activation of microglia following SAH, which will improve our ability to study the biology of microglial activation, and may ultimately improve management of disease progression and response to therapies directed at microglial activation.
Collapse
Affiliation(s)
- Benjamin A Plog
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA; Department of Pathology, University of Rochester Medical Center, Rochester, NY, USA.
| | - Katherine M Moll
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Hongyi Kang
- Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Jeffrey J Iliff
- Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Matthew L Dashnaw
- Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Maiken Nedergaard
- Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - G Edward Vates
- Department of Neurosurgery, Center for Translation Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|