1
|
Abdalkader M, Nguyen TN, Sahoo A, Qureshi MM, Ong CJ, Klein P, Miller MI, Mian AZ, Kaesmacher J, Mujanovic A, Hu W, Chen HS, Setty BN. Contrast Staining in Noninfarcted Tissue after Endovascular Treatment of Acute Ischemic Stroke. AJNR Am J Neuroradiol 2024; 45:701-707. [PMID: 38697792 PMCID: PMC11288587 DOI: 10.3174/ajnr.a8222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/03/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND AND PURPOSE Contrast staining is a common finding after endovascular treatment of acute ischemic stroke. It typically occurs in infarcted tissue and is considered an indicator of irreversible brain damage. Contrast staining in noninfarcted tissue has not been systematically investigated. We sought to assess the incidence, risk factors, and clinical significance of contrast staining in noninfarcted tissue after endovascular treatment. MATERIALS AND METHODS We conducted a retrospective review of consecutive patients who underwent endovascular treatment for anterior circulation large-vessel occlusion acute ischemic stroke. Contrast staining, defined as new hyperdensity on CT after endovascular treatment, was categorized as either contrast staining in infarcted tissue if the stained region demonstrated restricted diffusion on follow-up MR imaging or contrast staining in noninfarcted tissue if the stained region demonstrated no restricted diffusion. Baseline differences between patients with and without contrast staining in noninfarcted tissue were compared. Logistic regression was used to identify independent associations for contrast staining in noninfarcted tissue after endovascular treatment. RESULTS Among 194 patients who underwent endovascular treatment for large-vessel occlusion acute ischemic stroke and met the inclusion criteria, contrast staining in infarcted tissue was noted in 52/194 (26.8%) patients; contrast staining in noninfarcted tissue, in 26 (13.4%) patients. Both contrast staining in infarcted tissue and contrast staining in noninfarcted tissue were noted in 5.6% (11/194). Patients with contrast staining in noninfarcted tissue were found to have a higher likelihood of having an ASPECTS of 8-10, to be associated with contrast staining in infarcted tissue, and to achieve successful reperfusion compared with those without contrast staining in noninfarcted tissue. In contrast staining in noninfarcted tissue regions, the average attenuation was 40 HU, significantly lower than the contrast staining in infarcted tissue regions (53 HU). None of the patients with contrast staining in noninfarcted tissue had clinical worsening during their hospital stay. The median discharge mRS was significantly lower in patients with contrast staining in noninfarcted tissue than in those without (3 versus 4; P = .018). No independent predictors of contrast staining in noninfarcted tissue were found. CONCLUSIONS Contrast staining can be seen outside the infarcted tissue after endovascular treatment of acute ischemic stroke, likely attributable to the reversible disruption of the BBB in ischemic but not infarcted tissue. While generally benign, understanding its characteristics is important because it may mimic pathologic conditions such as infarcted tissue and cerebral edema.
Collapse
Affiliation(s)
- Mohamad Abdalkader
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Thanh N Nguyen
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology (T.N.N., C.J.O.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Anurag Sahoo
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Muhammad M Qureshi
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Charlene J Ong
- Department of Neurology (T.N.N., C.J.O.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
- Department of Neurology (C.J.O.), Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Piers Klein
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Matthew I Miller
- Department of Medicine (M.I.M.), Cambridge Health Alliance, Cambridge, Massachusetts
| | - Asim Z Mian
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| | - Johannes Kaesmacher
- Institute of Diagnostic and Interventional Neuroradiology, Institute of Diagnostic, Interventional and Pediatric Radiology and Department of Neurology (J.K., A.M.), Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Adnan Mujanovic
- Institute of Diagnostic and Interventional Neuroradiology, Institute of Diagnostic, Interventional and Pediatric Radiology and Department of Neurology (J.K., A.M.), Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Wei Hu
- Department of Neurology and Stroke Center (W.H.), Division of Life Sciences and Medicine, The First Affiliated Hospital of the University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Sheng Chen
- Department of Neurology (H.S.C.), General Hospital of Northern Theater Command, Shenyang, China
| | - Bindu N Setty
- From the Department of Radiology (M.A., T.N.N., A.S., M.M.Q., P.K., A.Z.M., B.N.S.), Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts
| |
Collapse
|
2
|
Davidson TL, Stevenson RJ. Vulnerability of the Hippocampus to Insults: Links to Blood-Brain Barrier Dysfunction. Int J Mol Sci 2024; 25:1991. [PMID: 38396670 PMCID: PMC10888241 DOI: 10.3390/ijms25041991] [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: 01/03/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The hippocampus is a critical brain substrate for learning and memory; events that harm the hippocampus can seriously impair mental and behavioral functioning. Hippocampal pathophysiologies have been identified as potential causes and effects of a remarkably diverse array of medical diseases, psychological disorders, and environmental sources of damage. It may be that the hippocampus is more vulnerable than other brain areas to insults that are related to these conditions. One purpose of this review is to assess the vulnerability of the hippocampus to the most prevalent types of insults in multiple biomedical domains (i.e., neuroactive pathogens, neurotoxins, neurological conditions, trauma, aging, neurodegenerative disease, acquired brain injury, mental health conditions, endocrine disorders, developmental disabilities, nutrition) and to evaluate whether these insults affect the hippocampus first and more prominently compared to other brain loci. A second purpose is to consider the role of hippocampal blood-brain barrier (BBB) breakdown in either causing or worsening the harmful effects of each insult. Recent research suggests that the hippocampal BBB is more fragile compared to other brain areas and may also be more prone to the disruption of the transport mechanisms that act to maintain the internal milieu. Moreover, a compromised BBB could be a factor that is common to many different types of insults. Our analysis indicates that the hippocampus is more vulnerable to insults compared to other parts of the brain, and that developing interventions that protect the hippocampal BBB may help to prevent or ameliorate the harmful effects of many insults on memory and cognition.
Collapse
Affiliation(s)
- Terry L. Davidson
- Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Avenue, NW, Washington, DC 20016, USA
| | | |
Collapse
|
3
|
Waseem A, Rashid S, Rashid K, Khan MA, Khan R, Haque R, Seth P, Raza SS. Insight into the transcription factors regulating Ischemic Stroke and Glioma in Response to Shared Stimuli. Semin Cancer Biol 2023; 92:102-127. [PMID: 37054904 DOI: 10.1016/j.semcancer.2023.04.006] [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: 11/23/2022] [Revised: 03/28/2023] [Accepted: 04/09/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemic stroke and glioma are the two leading causes of patient mortality globally. Despite physiological variations, 1 in 10 people who have an ischemic stroke go on to develop brain cancer, most notably gliomas. In addition, glioma treatments have also been shown to increase the risk of ischemic strokes. Stroke occurs more frequently in cancer patients than in the general population, according to traditional literature. Unbelievably, these events share multiple pathways, but the precise mechanism underlying their co-occurrence remains unknown. Transcription factors (TFs), the main components of gene expression programmes, finally determine the fate of cells and homeostasis. Both ischemic stroke and glioma exhibit aberrant expression of a large number of TFs, which are strongly linked to the pathophysiology and progression of both diseases. The precise genomic binding locations of TFs and how TF binding ultimately relates to transcriptional regulation remain elusive despite a strong interest in understanding how TFs regulate gene expression in both stroke and glioma. As a result, the importance of continuing efforts to understand TF-mediated gene regulation is highlighted in this review, along with some of the primary shared events in stroke and glioma.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| | - Sumaiya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Khalid Rashid
- Department of Cancer Biology, Vontz Center for Molecular Studies, Cincinnati, OH 45267-0521
| | | | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City,Mohali, Punjab 140306, India
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya -824236, India
| | - Pankaj Seth
- Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Haryana-122052, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| |
Collapse
|
4
|
Baek SJ, Hammock BD, Hwang IK, Li Q, Moustaid-Moussa N, Park Y, Safe S, Suh N, Yi SS, Zeldin DC, Zhong Q, Bradbury JA, Edin ML, Graves JP, Jung HY, Jung YH, Kim MB, Kim W, Lee J, Li H, Moon JS, Yoo ID, Yue Y, Lee JY, Han HJ. Natural Products in the Prevention of Metabolic Diseases: Lessons Learned from the 20th KAST Frontier Scientists Workshop. Nutrients 2021; 13:1881. [PMID: 34072678 PMCID: PMC8227583 DOI: 10.3390/nu13061881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
The incidence of metabolic and chronic diseases including cancer, obesity, inflammation-related diseases sharply increased in the 21st century. Major underlying causes for these diseases are inflammation and oxidative stress. Accordingly, natural products and their bioactive components are obvious therapeutic agents for these diseases, given their antioxidant and anti-inflammatory properties. Research in this area has been significantly expanded to include chemical identification of these compounds using advanced analytical techniques, determining their mechanism of action, food fortification and supplement development, and enhancing their bioavailability and bioactivity using nanotechnology. These timely topics were discussed at the 20th Frontier Scientists Workshop sponsored by the Korean Academy of Science and Technology, held at the University of Hawaii at Manoa on 23 November 2019. Scientists from South Korea and the U.S. shared their recent research under the overarching theme of Bioactive Compounds, Nanoparticles, and Disease Prevention. This review summarizes presentations at the workshop to provide current knowledge of the role of natural products in the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Seung J. Baek
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Bruce D. Hammock
- Department of Entomology, University of California, Davis, CA 95616, USA;
| | - In-Koo Hwang
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Qingxiao Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA;
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA;
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (Y.P.); (Y.Y.)
| | - Stephen Safe
- Department of Biochemistry & Biophysics, Texas A & M University, College Station, TX 77843, USA;
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Sun-Shin Yi
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Darryl C. Zeldin
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Qixin Zhong
- Department of Food Sciences, University of Tennessee, Knoxville, TN 37996, USA;
| | - Jennifer Alyce Bradbury
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Matthew L. Edin
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Joan P. Graves
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Hyo-Young Jung
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Young-Hyun Jung
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Mi-Bo Kim
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Woosuk Kim
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Jaehak Lee
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Hong Li
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Jong-Seok Moon
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Ik-Dong Yoo
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (Y.P.); (Y.Y.)
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Ho-Jae Han
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| |
Collapse
|
5
|
Ciacciarelli A, Sette G, Giubilei F, Orzi F. Chronic cerebral hypoperfusion: An undefined, relevant entity. J Clin Neurosci 2020; 73:8-12. [PMID: 31948882 DOI: 10.1016/j.jocn.2020.01.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/06/2020] [Indexed: 01/11/2023]
Abstract
Despite the large body of data available, chronic cerebral hypoperfusion lacks an operative definition. In a tautological way, the term hypoperfusion is being referred to conditions of "inadequate blood flow", "defects of perfusion" or "dysfunction of autoregulation". The chronicity refers to sustained conditions or wavering states characterized by repeated phases of inefficient functional hyperemia. The phenomenon may affect the whole brain or defined areas. A few defined clinical disorders, including heart failure, hypotension, atherosclerosis of large or small vessels and carotid stenosis are thought to cause progressive brain disorders due to chronic hypoperfusion. The clinical relevance manifests mostly as neurocognitive disorders associated with neuroimaging changes.The available data support a conceptual framework that considerschronic cerebral hypoperfusiona likely, relevant pathogenic mechanism for the neurodegeneration-like progression of the neurocognitive disorders. The relationship between neuropathology, cerebral perfusion, and symptoms progression is, however, elusive for several aspects. Typical microangiopathy findings, such as MRI white matter hyperintensities, may appear in individuals without any cerebrovascular risk or vascular lesions. Pathology features of the MRI changes, such as demyelination and gliosis, may result from dysfunction of the neuro-vascular unit not directly associated withvascular mechanisms. In this review, we aim to overview the most common clinical conditions thought to reflect chronic hypoperfusion.
Collapse
Affiliation(s)
- Antonio Ciacciarelli
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy.
| | - Giuliano Sette
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Franco Giubilei
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| | - Francesco Orzi
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), "SAPIENZA" University of Rome, Sant'Andrea University Hospital, Rome, Italy
| |
Collapse
|
6
|
Li J, Liu Y, Zhang X, Chen R, Zhang L, Xue J, Gao X. Dl-3-N-Butylphthalide Alleviates the Blood–Brain Barrier Permeability of Focal Cerebral Ischemia Reperfusion in Mice. Neuroscience 2019; 413:99-107. [DOI: 10.1016/j.neuroscience.2019.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/04/2019] [Accepted: 06/13/2019] [Indexed: 10/26/2022]
|
7
|
Cellular and Molecular Differences Between Area CA1 and the Dentate Gyrus of the Hippocampus. Mol Neurobiol 2019; 56:6566-6580. [PMID: 30874972 DOI: 10.1007/s12035-019-1541-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/27/2019] [Indexed: 12/16/2022]
Abstract
A distinct feature of the hippocampus of the brain is its unidirectional tri-synaptic pathway originating from the entorhinal cortex and projecting to the dentate gyrus (DG) then to area CA3 and subsequently, area CA1 of the Ammon's horn. Each of these areas of the hippocampus has its own cellular structure and distinctive function. The principal neurons in these areas are granule cells in the DG and pyramidal cells in the Ammon's horn's CA1 and CA3 areas with a vast network of interneurons. This review discusses the fundamental differences between the CA1 and DG areas regarding cell morphology, synaptic plasticity, signaling molecules, ability for neurogenesis, vulnerability to various insults and pathologies, and response to pharmacological agents.
Collapse
|
8
|
Ghosh S, Sarkar S, Choudhury ST, Ghosh T, Das N. Triphenyl phosphonium coated nano-quercetin for oral delivery: Neuroprotective effects in attenuating age related global moderate cerebral ischemia reperfusion injury in rats. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2439-2450. [PMID: 28822845 DOI: 10.1016/j.nano.2017.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/28/2017] [Accepted: 08/04/2017] [Indexed: 01/04/2023]
Abstract
Cerebral ischemia-reperfusion is a classic example of reactive oxygen species (ROS) mediated acute damage to brain. Post-ischemic reperfusion induced oxygen free radicals production causes damage to brain cell mitochondria. Antioxidants like quercetin (Qc) have potentials to manage oxidative stress related pathophysiology. However low oral bioavailability and poor cell membrane permeability restrict its therapeutic efficacy. To overcome these hurdles mitochondria specific delivery of Qc nanocapsules was designed to efficiently counteract cerebral ischemia-reperfusion induced cell death and neurodegeneration in young and aged rats. The orally deliverable quercetin loaded polymeric nanocapsules (N1QC) were made mitochondria specific by using triphenylphosphonium cation as one of the matrix components. N1QC demonstrated higher brain uptake and remarkable mitochondrial localization post cerebral ischemia-reperfusion. This unique controlled mitochondrial delivery of quercetin ameliorated histopathological severity by preserving mitochondrial structural and functional integrity through sequestering ROS thus modulating mitochondrial ROS mediated apoptotic cell death in young and aged rats.
Collapse
Affiliation(s)
- Swarupa Ghosh
- Drug Development, Diagnostics and Biotechnology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India.
| | - Sibani Sarkar
- Drug Development, Diagnostics and Biotechnology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Somsubhra T Choudhury
- Drug Development, Diagnostics and Biotechnology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Tirtha Ghosh
- SRSV (Govt. Polytechnic), P.O. Suri, Dist-, Birbhum, West Bengal, India
| | - Nirmalendu Das
- Drug Development, Diagnostics and Biotechnology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
9
|
Yoo DY, Lee KY, Park JH, Jung HY, Kim JW, Yoon YS, Won MH, Choi JH, Hwang IK. Glucose metabolism and neurogenesis in the gerbil hippocampus after transient forebrain ischemia. Neural Regen Res 2016; 11:1254-9. [PMID: 27651772 PMCID: PMC5020823 DOI: 10.4103/1673-5374.189189] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent evidence exists that glucose transporter 3 (GLUT3) plays an important role in the energy metabolism in the brain. Most previous studies have been conducted using focal or hypoxic ischemia models and have focused on changes in GLUT3 expression based on protein and mRNA levels rather than tissue levels. In the present study, we observed change in GLUT3 immunoreactivity in the adult gerbil hippocampus at various time points after 5 minutes of transient forebrain ischemia. In the sham-operated group, GLUT3 immunoreactivity in the hippocampal CA1 region was weak, in the pyramidal cells of the CA1 region increased in a time-dependent fashion 24 hours after ischemia, and in the hippocampal CA1 region decreased significantly between 2 and 5 days after ischemia, with high level of GLUT3 immunoreactivity observed in the CA1 region 10 days after ischemia. In a double immunofluorescence study using GLUT3 and glial-fibrillary acidic protein (GFAP), we observed strong GLUT3 immunoreactivity in the astrocytes. GLUT3 immunoreactivity increased after ischemia and peaked 7 days in the dentate gyrus after ischemia/reperfusion. In a double immunofluorescence study using GLUT3 and doublecortin (DCX), we observed low level of GLUT3 immunoreactivity in the differentiated neuroblasts of the subgranular zone of the dentate gyrus after ischemia. GLUT3 immunoreactivity in the sham-operated group was mainly detected in the subgranular zone of the dentate gyrus. These results suggest that the increase in GLUT3 immunoreactivity may be a compensatory mechanism to modulate glucose level in the hippocampal CA1 region and to promote adult neurogenesis in the dentate gyrus.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Kwon Young Lee
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
10
|
Chelluboina B, Klopfenstein JD, Pinson DM, Wang DZ, Vemuganti R, Veeravalli KK. Matrix Metalloproteinase-12 Induces Blood–Brain Barrier Damage After Focal Cerebral Ischemia. Stroke 2015; 46:3523-31. [DOI: 10.1161/strokeaha.115.011031] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/07/2015] [Indexed: 12/12/2022]
Abstract
Background and Purpose—
Matrix metalloproteinases (MMPs) have a central role in compromising the integrity of the blood–brain barrier (BBB). The role of MMP-12 in brain damage after ischemic stroke remains unknown. The main objective of the current study is to investigate the effect of MMP-12 suppression at an early time point before reperfusion on the BBB damage in rats.
Methods—
Sprague–Dawley rats were subjected to middle cerebral artery occlusion and reperfusion. MMP-12 shRNA–expressing plasmids formulated as nanoparticles were administered at a dose of 1 mg/kg body weight. The involvement of MMP-12 on BBB damage was assessed by performing various techniques, including Evans blue dye extravasation, 2,3,5-triphenyltetrazolium chloride staining, immunoblot, gelatin zymography, and immunofluorescence analysis.
Results—
MMP-12 is upregulated ≈31-, 47-, and 66-fold in rats subjected 1–, 2-, or 4-hour ischemia, respectively, followed by 1-day reperfusion. MMP-12 suppression protected the BBB integrity by inhibiting the degradation of tight-junction proteins. Either intravenous or intra-arterial delivery of MMP-12 shRNA-expressing plasmid significantly reduced the percent Evans blue dye extravasation and infarct size. Furthermore, MMP-12 suppression reduced the endogenous levels of other proteases, such as tissue-type plasminogen activator and MMP-9, which are also known to be the key players involved in BBB damage.
Conclusions—
These results demonstrate the adverse role of MMP-12 in acute brain damage that occurs after ischemic stroke and, thereby, suggesting that MMP-12 suppression could be a promising therapeutic target for cerebral ischemia.
Collapse
Affiliation(s)
- Bharath Chelluboina
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Jeffrey D. Klopfenstein
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - David M. Pinson
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - David Z. Wang
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Raghu Vemuganti
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| | - Krishna Kumar Veeravalli
- From the Departments of Cancer Biology and Pharmacology (B.C., K.K.V.), Neurosurgery (J.D.K.), Pathology (D.M.P.), and Neurology (D.Z.W.), University of Illinois College of Medicine at Peoria; Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison (R.V.); and Comprehensive Stroke Center, Illinois Neurological Institute, Peoria (J.D.K., D.Z.W.)
| |
Collapse
|
11
|
Delayed hyperoxic ventilation attenuates oxygen-induced free radical accumulation during early reperfusion after global brain ischemia. Neuroreport 2015; 26:131-8. [PMID: 25569795 DOI: 10.1097/wnr.0000000000000314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To compare the effect of immediate and delayed administration of oxygen on the accumulation of free radicals in ischemia-reperfusion animal models. Thirty-two adult male Mongolian gerbils with microdialysis probes implanted in the right hippocampal CA1 were divided randomly into four groups (eight each). One group was sham-operated (Sham group) whereas the other three groups were subjected to 10 min bilateral carotid artery occlusion (BCAO). BCAO-treated animals were then subjected to the following: (a) immediate 30% O2 (near normoxia, NO group), (b) immediate 100% O2 (hyperoxia, HO group), and (c) 30% O2 for 60 min, followed by 100% O2 for 60 min (delayed hyperoxia, DHO group). Hippocampal accumulation of hydroxyl radicals (•OH) during reperfusion was estimated by measuring 2,3-dihydroxybenzoic acid (DHBA) and 2,5-DHBA in microdialysis perfusate. Hippocampi were removed 2 h after perfusion to measure malondialdehyde, pyruvate dehydrogenase activity, indices of lipid peroxidation, and cellular respiration. At 24 h after BCAO, the histology of hippocampi was analyzed to rate the injury. Immediately after the onset of reperfusion, all groups showed markedly elevated DHBA, which returned to baseline over 1-2 h. Compared with the NO group, the HO group showed significantly higher peak DHBA and slower recovery. In contrast, the DHO group was not significantly different from the NO group in terms of the DHBA level. DHO animals also showed significantly lower hippocampal malondialdehyde accumulation and higher pyruvate dehydrogenase activity at 2 h after reperfusion versus the HO group. Histology analysis also showed animals in the DHO group with ameliorated injury compared with the HO group. Hydroxyl radical accumulation was more sensitive to O2 during early reperfusion. Delayed hyperoxia may re-establish oxidative metabolism while minimizing oxidative stress after CA.
Collapse
|
12
|
Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood-brain barrier. Nat Med 2013; 19:1584-96. [PMID: 24309662 DOI: 10.1038/nm.3407] [Citation(s) in RCA: 1718] [Impact Index Per Article: 143.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/22/2013] [Indexed: 01/01/2023]
Abstract
The interface between the blood circulation and the neural tissue features unique characteristics that are encompassed by the term 'blood-brain barrier' (BBB). The main functions of this barrier, namely maintenance of brain homeostasis, regulation of influx and efflux transport, and protection from harm, are determined by its specialized multicellular structure. Every constituent cell type makes an indispensable contribution to the BBB's integrity. But if one member of the BBB fails, and as a result the barrier breaks down, there can be dramatic consequences and neuroinflammation and neurodegeneration can occur. In this Review, we highlight recently gained mechanistic insights into the development and maintenance of the BBB. We then discuss how BBB disruption can cause or contribute to neurological disease. Finally, we examine how this knowledge can be used to explore new possibilities for BBB repair.
Collapse
Affiliation(s)
- Birgit Obermeier
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | |
Collapse
|
13
|
Huang S, Farrar CT, Dai G, Kwon SJ, Bogdanov AA, Rosen BR, Kim YR. Dynamic monitoring of blood-brain barrier integrity using water exchange index (WEI) during mannitol and CO2 challenges in mouse brain. NMR IN BIOMEDICINE 2013; 26:376-85. [PMID: 23055278 PMCID: PMC4029920 DOI: 10.1002/nbm.2871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 07/16/2012] [Accepted: 08/27/2012] [Indexed: 05/13/2023]
Abstract
The integrity of the blood-brain barrier (BBB) is critical to normal brain function. Traditional techniques for the assessment of BBB disruption rely heavily on the spatiotemporal analysis of extravasating contrast agents. However, such methods based on the leakage of relatively large molecules are not suitable for the detection of subtle BBB impairment or for the performance of repeated measurements in a short time frame. Quantification of the water exchange rate constant (WER) across the BBB using strictly intravascular contrast agents could provide a much more sensitive method for the quantification of the BBB integrity. To estimate WER, we have recently devised a powerful new method using a water exchange index (WEI) biomarker and demonstrated BBB disruption in an acute stroke model. Here, we confirm that WEI is sensitive to even very subtle changes in the integrity of the BBB caused by: (i) systemic hypercapnia and (ii) low doses of a hyperosmolar solution. In addition, we have examined the sensitivity and accuracy of WEI as a biomarker of WER using computer simulation. In particular, the dependence of the WEI-WER relation on changes in vascular blood volume, T1 relaxation of cellular magnetization and transcytolemmal water exchange was explored. Simulated WEI was found to vary linearly with WER for typically encountered exchange rate constants (1-4 Hz), regardless of the blood volume. However, for very high WER (>5 Hz), WEI became progressively more insensitive to increasing WER. The incorporation of transcytolemmal water exchange, using a three-compartment tissue model, helped to extend the linear WEI regime to slightly higher WER, but had no significant effect for most physiologically important WERs (WER < 4 Hz). Variation in cellular T1 had no effect on WEI. Using both theoretical and experimental approaches, our study validates the utility of the WEI biomarker for the monitoring of BBB integrity.
Collapse
Affiliation(s)
- Shuning Huang
- Health Science and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA 02139
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Guangping Dai
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Seon Joo Kwon
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | | | - Bruce R. Rosen
- Health Science and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA 02139
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Young R. Kim
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
- Corresponding author: Young Ro Kim Assistant Professor, Harvard Medical School Harvard-MIT (HST) Athinoula A. Martinos Center for Biomedical Imaging Massachusetts General Hospital-East Bldg. 149, 13th Street Charlestown, MA 02129 Fax: 617-726-7422
| |
Collapse
|
14
|
Chip S, Nitsch C, Wellmann S, Kapfhammer JP. Subfield-specific neurovascular remodeling in the entorhino-hippocampal-organotypic slice culture as a response to oxygen-glucose deprivation and excitotoxic cell death. J Cereb Blood Flow Metab 2013; 33:508-18. [PMID: 23232944 PMCID: PMC3618384 DOI: 10.1038/jcbfm.2012.190] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Transient ischemia causes delayed neurodegeneration in selective brain areas, particularly in the CA1 field of the hippocampus. This is accompanied by neurovascular impairment. It is unknown whether neurodegeneration is the cause or consequence of vascular changes. In an entorhino-hippocampal-organotypic slice culture system with well-preserved blood vessels, we studied the interplay between neurodegeneration and neurovasculature. Short-term oxygen and glucose deprivation (OGD) resulted in upregulation of hypoxic markers and with a delay of 24 to 48 hours in selective nerve cell death in CA1. In parallel, local vessel density decreased as detected by markers of endothelial cells and of the extracellular matrix. Claudin-5, a tight junction protein and marker of the blood-brain barrier was reduced. Preventing neuronal death with tetrodotoxin or 6-cyano-7-nitroquinoxaline-2,3-dione rescued blood vessels, suggesting that vessel loss is not due to OGD per se but a consequence of neuronal death. Induction of excitotoxic neuronal death with AMPA caused widespread neurodegeneration, but vessel reduction was confined to CA1. In dentate gyrus without neuronal loss, vessel density increased. We propose that neuronal stress and death influence maintenance, loss and remodeling of the neurovasculature and that the type of vascular response is in addition determined by local factors within the hippocampus.
Collapse
Affiliation(s)
- Sophorn Chip
- Department of Biomedicine, Section of Functional Neuroanatomy, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
15
|
Babu R, Bagley JH, Di C, Friedman AH, Adamson C. Thrombin and hemin as central factors in the mechanisms of intracerebral hemorrhage-induced secondary brain injury and as potential targets for intervention. Neurosurg Focus 2012; 32:E8. [PMID: 22463118 DOI: 10.3171/2012.1.focus11366] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stoke that may cause significant morbidity and mortality. Brain injury due to ICH initially occurs within the first few hours as a result of mass effect due to hematoma formation. However, there is increasing interest in the mechanisms of secondary brain injury as many patients continue to deteriorate clinically despite no signs of rehemorrhage or hematoma expansion. This continued insult after primary hemorrhage is believed to be mediated by the cytotoxic, excitotoxic, oxidative, and inflammatory effects of intraparenchymal blood. The main factors responsible for this injury are thrombin and erythrocyte contents such as hemoglobin. Therapies including thrombin inhibitors, N-methyl-D-aspartate antagonists, chelators to bind free iron, and antiinflammatory drugs are currently under investigation for reducing this secondary brain injury. This review will discuss the molecular mechanisms of brain injury as a result of intraparenchymal blood, potential targets for therapeutic intervention, and treatment strategies currently in development.
Collapse
Affiliation(s)
- Ranjith Babu
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | |
Collapse
|
16
|
Fukuhara A, Nakajima H, Miyamoto Y, Inoue K, Kume S, Lee YH, Noda M, Uchiyama S, Shimamoto S, Nishimura S, Ohkubo T, Goto Y, Takeuchi T, Inui T. Drug delivery system for poorly water-soluble compounds using lipocalin-type prostaglandin D synthase. J Control Release 2011; 159:143-50. [PMID: 22226778 DOI: 10.1016/j.jconrel.2011.12.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 11/15/2011] [Accepted: 12/19/2011] [Indexed: 10/14/2022]
Abstract
Lipocalin-type prostaglandin D synthase (L-PGDS) is a member of the lipocalin superfamily and a secretory lipid-transporter protein, which binds a wide variety of hydrophobic small molecules. Here we show the feasibility of a novel drug delivery system (DDS), utilizing L-PGDS, for poorly water-soluble compounds such as diazepam (DZP), a major benzodiazepine anxiolytic drug, and 6-nitro-7-sulfamoylbenzo[f]quinoxaline-2,3-dione (NBQX), an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist and anticonvulsant. Calorimetric experiments revealed for both compounds that each L-PGDS held three molecules with high binding affinities. By mass spectrometry, the 1:3 complex of L-PGDS and NBQX was observed. L-PGDS of 500μM increased the solubility of DZP and NBQX 7- and 2-fold, respectively, compared to PBS alone. To validate the potential of L-PGDS as a drug delivery vehicle in vivo, we have proved the prospective effects of these compounds via two separate delivery strategies. First, the oral administration of a DZP/L-PGDS complex in mice revealed an increased duration of pentobarbital-induced loss of righting reflex. Second, the intravenous treatment of ischemic gerbils with NBQX/L-PGDS complex showed a protective effect on delayed neuronal cell death at the hippocampal CA1 region. We propose that our novel DDS could facilitate pharmaceutical development and clinical usage of various water-insoluble compounds.
Collapse
Affiliation(s)
- Ayano Fukuhara
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Ghosh S, Das N, Mandal AK, Dungdung SR, Sarkar S. Mannosylated liposomal cytidine 5' diphosphocholine prevent age related global moderate cerebral ischemia reperfusion induced mitochondrial cytochrome c release in aged rat brain. Neuroscience 2010; 171:1287-99. [PMID: 20883746 DOI: 10.1016/j.neuroscience.2010.09.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 09/18/2010] [Accepted: 09/23/2010] [Indexed: 10/19/2022]
Abstract
Mitochondrial dysfunctions generating from cerebral ischemia-reperfusion exert a potential threat on neuronal cell survival and hence, accelerate the aging process and age dependent neuropathology. Thirty min moderate cerebral ischemia induced by bilateral common carotid artery occlusion (BCCAO) followed by 30 min reperfusion caused an increased diene production, depleted glutathione (GSH) content, reduced superoxide dismutase (SOD) and catalase activities and pyramidal neuronal loss in young (2 months old) and aged (20 months old) rat brain compared to sham operated controls. Cytidine 5' diphosphocholine (CDP-Choline) is a known neuroprotective drug. CDP-Choline after metabolism in the liver suffers hydrolysis and splits into cytidine and choline before entering systemic circulation and hardly circumvents blood brain barrier (BBB) as such. Previous reports show CDP-Choline liposomes significantly increased in vivo uptake compared to "free drug" administration in cerebral ischemia. To enhance the therapeutic concentration build up in brain we sought to formulate mannosylated liposomal CDP-Choline (MLCDP) utilizing the mannose receptors. We tested the therapeutic supremacy of MLCDP over liposomal CDP-Choline (LCDP) in global moderate cerebral ischemia reperfusion induced neuronal damage. CDP-Choline in MLCDP delivery system was found potent to exert substantial protection against global moderate cerebral ischemia reperfusion induced mitochondrial damage in aged rat brain. Membrane lipid peroxidation, GSSG/GSH ratio and reactive oxygen species (ROS) generation in cerebral tissue were found to be higher in aged, compared to young rat. Further decline of those parameters was observed in aged rat brain by the induction of global moderate cerebral ischemia and reperfusion. MLCDP treatment when compared to free or LCDP treatment prevented global moderate cerebral ischemia-reperfusion induced mitochondrial damage as evident ultra structurally and release of cytochrome c (cyt c) from mitochondria into cytosol and protected mitochondria to restore its normal structure and functions.
Collapse
Affiliation(s)
- S Ghosh
- Biomembrane Division, Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Kolkata-700032, India
| | | | | | | | | |
Collapse
|
18
|
Donzelli A, Braida D, Finardi A, Capurro V, Valsecchi AE, Colleoni M, Sala M. Neuroprotective Effects of Genistein in Mongolian Gerbils: Estrogen Receptor–β Involvement. J Pharmacol Sci 2010; 114:158-67. [DOI: 10.1254/jphs.10164fp] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
19
|
Hagberg H, Mallard C, Rousset CI, Wang X. Apoptotic mechanisms in the immature brain: involvement of mitochondria. J Child Neurol 2009; 24:1141-6. [PMID: 19574577 PMCID: PMC3674552 DOI: 10.1177/0883073809338212] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Brain injury after hypoxic-ischemic encephalopathy often develops with delayed appearance, opening a therapeutic window. Clinical studies in newborns show that post-hypoxic-ischemic hypothermia improves outcome. This has generated renewed interest in the molecular mechanisms of hypoxic-ischemic brain injury. In this brief review, we propose that mitochondrial permeabilization is crucial for injury to advance beyond the point of no return. We suggest that excitatory amino acids, nitric oxide, inflammation, trophic factor withdrawal, and an increased pro- versus antiapoptotic Bcl-2 protein ratio will trigger Bax-dependent mitochondrial outer membrane permeabilization. Mitochondrial outer membrane permeabilization, in turn, elicits mitochondrial release of cytochrome C, apoptosis-inducing factor, second mitochondria-derived activator of caspase/Diablo, and HtrA2/Omi. Cytochrome C efflux activates caspase-9/-3, leading to DNA fragmentation. Apoptosis-inducing factor interacts with cyclophilin A and induces chromatinolysis. Blockage of mitochondrial outer membrane permeabilization holds promise as a strategy for perinatal brain protection.
Collapse
Affiliation(s)
- Henrik Hagberg
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, Göteborg University, Sweden.
| | | | | | | |
Collapse
|
20
|
Castri P, Busceti C, Battaglia G, Girardi F, Cavallari M, Orzi F, Fornai F. Protection by Apomorphine in Two Independent Models of Acute Inhibition of Oxidative Metabolism in Rodents. Clin Exp Hypertens 2009; 28:387-94. [PMID: 16833051 DOI: 10.1080/10641960600549827] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Apomorphine was administered by continuous infusion in the mouse following acute inhibition of oxidative metabolism induced by systemic administration of MPTP, and in the gerbil following transient occlusion of the carotid arteries. The dosage employed was comparable to the one used in the treatment of severe on-off fluctuations in Parkinson's disease. The results show that apomorphine significantly diminishes the striatal lesion caused by MPTP and the size of the infarct associated with the transient global ischemia. These data suggest that apomorphine is neuroprotective, probably by means of an antioxidant effect, at doses that are clinically used. The finding may be relevant to brain ischemia as well to chronic neurodegeneration.
Collapse
Affiliation(s)
- Paola Castri
- Department of Neurological Sciences, University of Roma, "La Sapienza", Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
21
|
Hossmann KA. Cerebral ischemia: Models, methods and outcomes. Neuropharmacology 2008; 55:257-70. [DOI: 10.1016/j.neuropharm.2007.12.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 12/06/2007] [Accepted: 12/10/2007] [Indexed: 01/31/2023]
|
22
|
Zani A, Braida D, Capurro V, Sala M. Delta9-tetrahydrocannabinol (THC) and AM 404 protect against cerebral ischaemia in gerbils through a mechanism involving cannabinoid and opioid receptors. Br J Pharmacol 2007; 152:1301-11. [PMID: 17965746 PMCID: PMC2189998 DOI: 10.1038/sj.bjp.0707514] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 09/07/2007] [Accepted: 09/18/2007] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE It has been suggested that the endocannabinoid system elicits neuroprotection against excitotoxic brain damage. In the present study the therapeutic potential of AM 404 on ischaemia-induced neuronal injury was investigated in vivo and compared with that of the classical cannabinoid receptor type 1 (CB1) agonist, delta 9-tetraydrocannabinol (THC), using a model of transient global cerebral ischaemia in the gerbil. EXPERIMENTAL APPROACH The effects of AM 404 (0.015-2 mg kg(-1)) and THC (0.05-2 mg kg(-1)), given 5 min after ischaemia, were measured from 1 h to 7 days in terms of electroencephalographic (EEG) total spectral power, spontaneous motor activity, memory function, rectal temperature and hippocampal CA1 neuronal count. KEY RESULTS Over the dose range tested, AM 404 (2 mg kg(-1)) and THC (1 mg kg(-1)) completely reversed the ischaemia-induced behavioural, EEG and histological damage. Only THC (1 and 2 mg kg(-1)) induced a decrease of body temperature. Pretreatment with the selective CB1 receptor antagonist, AM 251 (1 mg kg(-1)) and the opioid antagonist, naloxone (2 mg kg(-1)) reversed the protective effect induced by both AM 404 and THC while the TRPV1 vanilloid antagonist, capsazepine (0.01 mg kg(-1)), was ineffective. CONCLUSIONS AND IMPLICATIONS Our findings demonstrate that AM 404 and THC reduce neuronal damage caused by bilateral carotid occlusion in gerbils and that this protection is mediated through an interaction with CB1 and opioid receptors. Endocannabinoids might form the basis for the development of new neuroprotective drugs useful for the treatment of stroke and other neurodegenerative pathologies.
Collapse
Affiliation(s)
- A Zani
- Department of Pharmacology, Chemotherapy and Medical Toxicology, Faculty of Sciences, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
23
|
Bahcekapili N, Uzüm G, Gökkusu C, Kuru A, Ziylan YZ. The relationship between erythropoietin pretreatment with blood-brain barrier and lipid peroxidation after ischemia/reperfusion in rats. Life Sci 2007; 80:1245-51. [PMID: 17300815 DOI: 10.1016/j.lfs.2006.12.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 12/04/2006] [Accepted: 12/06/2006] [Indexed: 10/23/2022]
Abstract
Blood-brain barrier (BBB) leakage plays a role in the pathogenesis of many pathological states of the brain including ischemia and some neurodegenerative disorders. In recent years, erythropoietin (EPO) has been shown to exert neuroprotection in many pathological conditions including ischemia in the brain. This study aimed to investigate the effects of EPO on BBB integrity, infarct size and lipid peroxidation following global brain ischemia/reperfusion in rats. Wistar male rats were divided into four groups (each group n=8); Group I; control group (sham-operated), Group II; ischemia/reperfusion group, Group III; EPO treated group (24 h before decapitation--000 U/kg r-Hu EPO i.p.), Group IV; EPO+ ischemia/reperfusion group (24 h before ischemia/reperfusion--3000 U/kg r-Hu EPO i.p.). Global brain ischemia was produced by the combination of bilateral common carotid arteries occlusion and hemorrhagic hypotension. Macroscopical and spectrophotometrical measurement of Evans Blue (EB) leakage was observed for BBB integrity. Infarct size was calculated based on 2,3,5-triphenyltetrazolium chloride (TTC) staining. Lipid peroxidation in the brain tissue was determined as the concentration of thiobarbituric acid-reactive substances (TBARS) for each group. Ischemic insult caused bilateral and regional BBB breakdown (hippocampus, cortex, corpus striatum, midbrain, brain stem and thalamus). EPO pretreatment reduced BBB disruption, infarct size and lipid peroxide levels in brain tissue with 20 min ischemia and 20 min reperfusion. These results suggest that EPO plays an important role in protecting against brain ischemia/reperfusion through inhibiting lipid peroxidation and decreasing BBB disruption.
Collapse
Affiliation(s)
- Nesrin Bahcekapili
- Istanbul University, Istanbul Faculty of Medicine, Department of Physiology, 34093, Capa Istanbul, Turkey
| | | | | | | | | |
Collapse
|
24
|
Pegorini S, Zani A, Braida D, Guerini-Rocco C, Sala M. Vanilloid VR1 receptor is involved in rimonabant-induced neuroprotection. Br J Pharmacol 2006; 147:552-9. [PMID: 16444289 PMCID: PMC1616983 DOI: 10.1038/sj.bjp.0706656] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Recently, a potential neuroprotective effect of rimonabant, independent of the CB1 receptor interaction, has been proposed. In the present study, the role of transient receptor potential channel vanilloid subfamily member 1, named VR1, on neuroprotective effect of rimonabant, on global cerebral ischemia in gerbils, was investigated. Rimonabant (0.05-3 mg kg-1), given i.p. 5 min after recirculation, dose dependently antagonized the ischemia-induced decrease in electroencephalographic (EEG) total spectral power and restored relative frequency band distribution 7 days after ischemia. Rimonabant (0.125-0.5 mg kg-1) fully prevented ischemia-induced hyperlocomotion 1 day after ischemia and memory impairment evaluated in a passive avoidance task, 3 days after ischemia. At 7 days after ischemia, the survival of pyramidal cells, in the CA1 subfield, was respectively 91 and 96%, in the animals given rimonabant 0.25 and 0.5 mg kg-1, compared to the vehicle group. Higher doses were not protective. The protection induced by rimonabant followed a bell-shaped curve, the maximal active doses being 0.25 and 0.5 mg kg-1. Capsazepine (0.01 mg kg-1), a selective VR1 vanilloid receptor antagonist, completely reversed rimonabant-induced neuroprotective effects against EEG flattening, memory impairment and CA1 hippocampal neuronal loss. These findings suggest that VR1 vanilloid receptors are involved in rimonabant's neuroprotection even if other mechanisms can contribute to this effect.
Collapse
Affiliation(s)
- Simona Pegorini
- Department of Pharmacology, Chemotherapy and Medical Toxicology, Faculty of Sciences, University of Milan, Via Vanvitelli 32, Milan 20129, Italy
| | - Alessia Zani
- Department of Pharmacology, Chemotherapy and Medical Toxicology, Faculty of Sciences, University of Milan, Via Vanvitelli 32, Milan 20129, Italy
| | - Daniela Braida
- Department of Pharmacology, Chemotherapy and Medical Toxicology, Faculty of Sciences, University of Milan, Via Vanvitelli 32, Milan 20129, Italy
| | - Chiara Guerini-Rocco
- Department of Pharmacology, Chemotherapy and Medical Toxicology, Faculty of Sciences, University of Milan, Via Vanvitelli 32, Milan 20129, Italy
| | - Mariaelvina Sala
- Department of Pharmacology, Chemotherapy and Medical Toxicology, Faculty of Sciences, University of Milan, Via Vanvitelli 32, Milan 20129, Italy
- Author for correspondence:
| |
Collapse
|
25
|
Sarkar S, Das N. Mannosylated liposomal flavonoid in combating age-related ischemia–reperfusion induced oxidative damage in rat brain. Mech Ageing Dev 2006; 127:391-7. [PMID: 16480758 DOI: 10.1016/j.mad.2005.12.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2005] [Revised: 12/28/2005] [Accepted: 12/28/2005] [Indexed: 12/21/2022]
Abstract
Active oxygen species alter the activities of the enzymes involved in the defence against free radicals and substantially influence the aging process and age-dependent neuropathology. Unilamellar liposomes were used to deliver flavonoidal antioxidant quercetin (QC) to rat brain. Antioxidant potential of QC loaded in mannosylated (QC 7.2 micromol/kg b.wt.) liposomes (50 nm) was investigated by an in vivo model of cerebral ischemia and reperfusion on Sprague Dawley young (2 months old, b.wt. 160-180 g) and aged (20 months old, b.wt. 415-440 g) rats. Animals were made ischemic for 30 min by bilateral clamping of the common carotid artery followed by a 30 min cerebral reperfusion by withdrawing the clamping. Diene level and (GSSG/GSH) ratio were found to be higher in normal aged, compared to normal young rat brain. Superoxide dismutase, catalase, glucose-6-phosphate dehydrogenase, glutathione reductase and glutathione S-transferase activities were lower in normal aged rat brain. Further reduction of these antioxidant enzymes was observed in aged rat brain by the induction of cerebral ischemia and reperfusion. Mannosylated liposomally encapsulated QC treatment resulted in a significant preservation of the activities of antioxidant enzymes and a marked inhibition of cellular edema formation in neuronal cells of young and old rats.
Collapse
Affiliation(s)
- Sibani Sarkar
- Biomembrane Division, Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | | |
Collapse
|
26
|
Weiskopf RB, Webb M, Stangle D, Klinbergs G, Toy P. A procedure for rapid issue of red cells for emergency use. Arch Pathol Lab Med 2005; 129:492-6. [PMID: 15794672 DOI: 10.5858/2005-129-492-apfrio] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT A College of American Pathologists Q-Probe revealed that the median turnaround times for emergency requests for red blood cells from the operating room were 30 minutes to release of cells from the blood bank and 34 minutes to delivery to the operating room. These times may not be adequate to permit the red cells to provide sufficiently rapid delivery of oxygen in massively bleeding patients. OBJECTIVE To improve the time from emergency request for red cells to delivery to the operating room. DESIGN A new emergency issue program was implemented for only the operating rooms; emergency issue to all other hospital locations remained unchanged. Six units of group O Rh-negative red blood cells (RBCs) are maintained in the blood bank in a separate basket with transfusion forms containing the unit numbers and expiration dates and a bag with one blood tubing segment from each unit. The times to issue and to delivery to the operating room suite were compared with time to issue of 2 group O Rh-negative RBCs for other hospital locations using the older system during the same time period and with the time to issue of 2 units to all other hospital locations during the preceding 2 years. SETTING A university hospital. MAIN OUTCOME MEASURES Time between emergency request for red cells and delivery to the operating room. RESULTS The time between blood bank notification and arrival in the operating room of the 6 units of RBCs was significantly shorter than the time required to just issue (not including delivery time) 2 units of RBCs to other hospital locations. With the new procedure, 82% of units issued reached the operating room within 2 minutes of request, 91% arrived within 3 minutes, and 100% arrived within 4 minutes. These percentages are significantly higher than those for only issue of blood (without delivery) using the older issuing procedure for all hospital locations during the previous 2 years (37%, 49%, and 66%, respectively; P = .007, .009, and .02, respectively) and for other locations during the same 7-month period (29%, 46%, and 73%, respectively; P = .004, .01, and .09, respectively). Time (mean [95% confidence interval]) from blood bank notification to delivery of RBCs to the operating room suite (2.1 [1.6-2.6] minutes, of which approximately 50-60 seconds is attributable to delivery time) was less than issue times (not including delivery times) using the older issuing procedure for other hospital locations during the same period (4.1 [3.1-5.0] minutes; P = .007). CONCLUSIONS An emergency issue procedure can be used to issue several units of RBCs within 1 minute and have them delivered to the operating room within 2 minutes while maintaining sufficient controls and providing required information to satisfy patient and blood bank requirements.
Collapse
Affiliation(s)
- Richard B Weiskopf
- Department of Anesthesia and Physiology, University of California, San Francisco 94143-0648, USA.
| | | | | | | | | |
Collapse
|
27
|
Pegorini S, Braida D, Verzoni C, Guerini-Rocco C, Consalez GG, Croci L, Sala M. Capsaicin exhibits neuroprotective effects in a model of transient global cerebral ischemia in Mongolian gerbils. Br J Pharmacol 2005; 144:727-35. [PMID: 15678080 PMCID: PMC1576053 DOI: 10.1038/sj.bjp.0706115] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Capsaicin, the irritant principle of hot peppers, is a vanilloid agonist known to activate the transient receptor potential channel vanilloid subfamily member 1 (VR1), recently reported to be involved in neurodegeneration. The present study investigated the role of VR1 in a model of global cerebral ischemia in gerbils. 2. Over the dose range tested, capsaicin (0.01, 0.025, 0.05, 0.2 and 0.6 mg kg(-1)), given 5 min after recirculation, dose-dependently antagonized the ischemia-induced electroencephalographic total spectral power decrease and restored relative frequency band distribution evaluated 7 days after ischemia. 3. Capsaicin, at all tested doses, fully prevented ischemia-induced hyperlocomotion evaluated 1 day after ischemia. 4. Capsaicin dose-dependently antagonized ischemia-induced memory impairment evaluated in a passive avoidance task, 3 days after ischemia. 5. Capsaicin showed a dose-dependent hypothermic effect evaluated for 2 h after recirculation. 6. At 7 days after ischemia, a progressive survival of pyramidal cells in the CA1 subfield in capsaicin-treated gerbils, with a maximum of 80%, at a dose of 0.2 mg kg(-1), was obtained. 7. The selective VR1 antagonist, capsazepine (0.01 mg kg(-1)), reversed capsaicin-induced protective effects, in a competitive manner. 8. These results suggest that the neuroprotective effect of capsaicin may be attributable, at least in part, to VR1 desensitization and provide a valuable target for development of interventional pharmacological strategies.
Collapse
Affiliation(s)
- Simona Pegorini
- Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milan, Via Vanvitelli 32, 20129 Milan, Italy
| | - Daniela Braida
- Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milan, Via Vanvitelli 32, 20129 Milan, Italy
| | - Chiara Verzoni
- Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milan, Via Vanvitelli 32, 20129 Milan, Italy
| | - Chiara Guerini-Rocco
- Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milan, Via Vanvitelli 32, 20129 Milan, Italy
| | | | - Laura Croci
- San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Mariaelvina Sala
- Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milan, Via Vanvitelli 32, 20129 Milan, Italy
- Author for correspondence:
| |
Collapse
|
28
|
Melgar MA, Rafols J, Gloss D, Diaz FG. Postischemic reperfusion: ultrastructural blood-brain barrier and hemodynamic correlative changes in an awake model of transient forebrain ischemia. Neurosurgery 2005; 56:571-81. [PMID: 15730583 DOI: 10.1227/01.neu.0000154702.23664.3d] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2004] [Accepted: 12/13/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In nonrecovery models of cerebral ischemia, blood-brain barrier (BBB) and cerebral blood flow (CBF) changes are known to occur during reperfusion. It is unknown, however, whether those CBF and BBB alterations occur after brief, transient ischemia with neurological recovery. The purpose of this study was to characterize the time course of CBF and BBB ultrastructural changes during reperfusion in an awake, recovery model of transient global forebrain ischemia (GFI). METHODS Forty-five adult Sprague-Dawley rats were subjected, while awake, to 10 minutes of GFI by the nine-vessel occlusion method. Thirty-five age-matched animals composed a sham-operated group. Normal control (n = 5), sham-operated (n = 5), and nine-vessel occlusion/reperfusion (n = 15) rats were selected for ultrastructural analysis. Electroencephalography was performed, and CBF, mean arterial blood pressure, and intracranial pressure were measured during ischemia and reperfusion up to 24 hours. Quantitative morphological analysis of cortical BBB capillaries was performed by transmission electron microscopy at the same time points at which specific CBF changes occurred during reperfusion. RESULTS CBF decreased to 6% of preocclusion values during GFI. This correlated with coma and decerebrate rigidity. During reperfusion, short-lived hyperemia (225 +/- 18%, P < 0.001) was characterized by increased intracranial pressure (28.3 +/- 2.6 mm Hg, P < 0.001) and isoelectric electroencephalogram. This was followed by hypoperfusion, which reached a nadir of 59.7% (59.7 +/- 8.8%, P < 0.01) from baseline by 90 minutes. At this time point, the electroencephalogram recovered, and intracranial pressure and mean arterial blood pressure showed no abnormalities. By 8.5 hours, CBF returned to normal, and this coincided with complete recovery of the animal. Ultrastructural BBB analysis revealed astrocyte end-foot process edema and patent capillaries during hyperemia. Severe interstitial BBB edema and capillary lumen collapse was observed during hypoperfusion. Detachment and migration of pericytes was observed during hypoperfusion and beyond. CONCLUSION A biphasic CBF response is elicited during reperfusion after brief nonlethal GFI under awake conditions.
Collapse
Affiliation(s)
- Miguel A Melgar
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA.
| | | | | | | |
Collapse
|
29
|
Herrmann M, Stern M, Vollenweider F, Nitsch C. Effect of inherent epileptic seizures on brain injury after transient cerebral ischemia in Mongolian gerbils. Exp Brain Res 2003; 154:176-82. [PMID: 14557906 DOI: 10.1007/s00221-003-1655-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2003] [Accepted: 07/30/2003] [Indexed: 10/26/2022]
Abstract
Subthreshold excitotoxic stimuli such as brief cerebral ischemia or chemically induced seizures modulate brain injury resulting from subsequent transient ischemia. Depending on the delay between the two insults, either tolerance or cumulative damage will develop. We were interested whether non-chemically induced inherent epileptic seizures as they occur in Mongolian gerbils have an effect on the outcome of a transient global ischemia, i.e., whether they are an interfering variable in ischemia experiments. Occurrence of spontaneous seizures in adult male gerbils was registered with a video-controlled seizure monitoring system. Bilateral occlusion of common carotid arteries was carried out 2 h or 24 h after the last generalized seizure. After 4 days survival, the extent of ischemia-induced neuronal damage and glial activation were assessed in the hippocampus and striatum. No significant difference in the ischemia induced nerve cell loss was observed in cresyl violet stained sections between the 2-h or 24-h interval gerbils. Neuronal expression of endothelial nitric oxide synthase in CA1 disappeared with neuronal degeneration. Distribution and degree of upregulation of glial fibrillary acidic protein as marker for astrocytes did not differ between the two groups. We concluded that non-chemically induced inherent epileptic seizures neither protect the gerbil brain from injury nor augment the degree of damage resulting from transient forebrain ischemia. Thus, inherent epileptic seizures do not influence the outcome of the insult, making the gerbil a reliable model for studies on transient brain ischemia.
Collapse
Affiliation(s)
- Martina Herrmann
- Section of Neuroanatomy, Institute of Anatomy, University of Basel, Pestalozzistr. 20, 4056 Basel, Switzerland
| | | | | | | |
Collapse
|
30
|
Krep H, Böttiger BW, Bock C, Kerskens CM, Radermacher B, Fischer M, Hoehn M, Hossmann KA. Time course of circulatory and metabolic recovery of cat brain after cardiac arrest assessed by perfusion- and diffusion-weighted imaging and MR-spectroscopy. Resuscitation 2003; 58:337-48. [PMID: 12969612 DOI: 10.1016/s0300-9572(03)00151-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Brain recovery after cardiac arrest (CA) was assessed in cats using arterial spin tagging perfusion-weighted imaging (PWI), diffusion-weighted imaging (DWI), and 1H-spectroscopy (1H-MRS). Cerebral reperfusion and metabolic recovery was monitored in the cortex and in basal ganglia for 6 h after cardiopulmonary resuscitation (CPR). Furthermore, the effects of an hypertonic/hyperoncotic solution (7.5% NaCl/6% hydroxyl ethyl starch, HES) and a tissue-type plasminogen activator (TPA), applied during CPR, were assessed on brain recovery. CA and CPR were carried out in the MR scanner by remote control. CA for 15-20 min was induced by electrical fibrillation of the heart, followed by CPR using a pneumatic vest. PWI after successful CPR revealed initial cerebral hyperperfusion followed by delayed hypoperfusion. Initial cerebral recirculation was improved after osmotic treatment. Osmotic and thrombolytic therapy were ineffective in ameliorating delayed hypoperfusion. Calculation of the apparent diffusion coefficient (ADC) from DWI demonstrated complete recovery of ion and water homeostasis in all animals. 1H-MRS measurements of lactate suggested an extended preservation of post-ischaemic anaerobic metabolism after TPA treatment. The combination of noninvasive MR techniques is a powerful tool for the evaluation of therapeutical strategies on circulatory and metabolic cerebral recovery after experimental cerebral ischaemia.
Collapse
Affiliation(s)
- Henning Krep
- Department of Anesthesia and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Krep H, Fischer M, Hoeft A. Endothelin-1 elevates regional cerebral perfusion during prolonged ventricular fibrillation cardiac arrest in pigs. Resuscitation 2003; 57:317-8. [PMID: 12804812 DOI: 10.1016/s0300-9572(03)00076-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
32
|
Iqbal S, Baziany A, Gordon S, Wright S, Hussain M, Miyashita H, Shuaib A, Hasan Rajput A. Neuroprotective effect of tiagabine in transient forebrain global ischemia: an in vivo microdialysis, behavioral, and histological study. Brain Res 2002; 946:162-70. [PMID: 12137918 DOI: 10.1016/s0006-8993(02)02871-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The neuroprotective effect of tiagabine was investigated in global ischemia in gerbils. Two groups of the animals received 15 mg/kg of tiagabine 30 min before ischemia. In the first group, the temperature was controlled at 37 degrees C from time of injection to 1 h after ischemia. In the second group, the temperature was left uncontrolled to see the hypothermic effect of tiagabine. Microdialysis was performed in CA1 region of hippocampus in half of the animals in each group to assess the levels of glutamate and gamma-amino-butyric acid (GABA). Animal behavior was also tested in 28-day groups in a radial-arm maze. Histology was done 7 and 28 days after ischemia in CA1 region of hippocampus to assess early and delayed effect of drug. A significant suppression of glutamate was noted in both groups (P<0.01). Behavioral results showed that in the temperature-uncontrolled treatment group, animals significantly reduced their working memory errors as compared to the temperature-controlled treatment group. Histology revealed a significant neuroprotection (P<0.001) in the temperature-uncontrolled treatment group. In the temperature-controlled treatment group, however, neuroprotection was insignificant (P>0.05). A third group of animals received the same dose of tiagabine 3 h after ischemia. Temperature was not controlled in this group. The animals were sacrificed after 7 days so no behavior testing was carried out. Histology showed no neuroprotection in this group (P>0.05). These results show that tiagabine offers a significant neuroprotection in global ischemia in gerbils when given 30 min before ischemia but not when given 3 h after ischemia.
Collapse
Affiliation(s)
- Saeeda Iqbal
- Department of Neurology, Royal University Hospital, Saskatoon, Saskatchewan, Canada S7N 0X8
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Hotta H, Uchida S, Kagitani F. Effects of stimulating the nucleus basalis of Meynert on blood flow and delayed neuronal death following transient ischemia in the rat cerebral cortex. THE JAPANESE JOURNAL OF PHYSIOLOGY 2002; 52:383-93. [PMID: 12519473 DOI: 10.2170/jjphysiol.52.383] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
An increase in cortical cerebral blood flow (CBF), independent of metabolic vasodilation, via the activation of cholinergic neurons originating in the nucleus basalis of Meynert (NBM) in the basal forebrain and projecting to the widespread cortices was recently demonstrated. In the present study, we aimed to clarify whether the increase in CBF following a stimulation of the NBM can improve delayed death of the cortical neurons following transient ischemia in rats. CBF was measured with a laser Doppler flowmeter, and the delayed neuronal death of the cerebral cortex produced by intermittent (every 5 s) occlusions of the unilateral common carotid artery for 60 min was measured histologically in the cortical hemisphere at 3 different coronal levels (6 microm thickness). In control rats without occlusion there were 6000-8000 intact neurons and 9-19 damaged neurons in the cortical hemisphere at each coronal level. During the occlusions, CBF ipsilateral to the occluded artery decreased by 13-32% of the preocclusion level. Five days after the occlusions, the numbers of damaged neurons were increased to 75-181. Repetitive electrical stimulation was delivered to the NBM, ipsilateral to the occluded artery, starting 5 min before the occlusions and finishing around the end of them. The increase in CBF induced by NBM stimulation prevented the occlusion-induced decrease in CBF in all 3 of the cortices. The delayed death of the cortical neurons previously observed after the occlusions was scarcely observable in all the cortices when NBM was stimulated. The present results suggest that NBM-originating vasodilative activation can protect the ischemia-induced delayed death of cortical neurons by preventing a blood flow decrease in widespread cortices.
Collapse
Affiliation(s)
- Harumi Hotta
- Motor and Autonomic Nervous System Integration Research Group, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015 Japan.
| | | | | |
Collapse
|
34
|
Yoneda Y, Mimura T, Kawagoe K, Yasukouchi T, Tatematu T, Ito M, Saito M, Sugimura M, Kito F, Kawajiri S. Discovery of diaminobutane derivatives as Ca(2+)-permeable AMPA receptor antagonists. Bioorg Med Chem 2002; 10:1347-59. [PMID: 11886798 DOI: 10.1016/s0968-0896(01)00398-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We designed and synthesized a series of the polyamine derivatives as potent Ca(2+)-permeable AMPA receptor antagonists. In the course of this study, we found that the polyamine derivatives exhibited strong hypotensive activity which was undesirable activity for neuroprotective agents. Therefore, we tried to find non-hypotensive antagonists by structural modification of such compounds. Through this derivatization, we obtained the diamine compounds having desired profiles. Especially, compound 8f, which was non-hypotensive and potent Ca(2+)-permeable AMPA receptor antagonist, showed neuroprotective effects in transient global ischemia models in gerbils.
Collapse
Affiliation(s)
- Yoshiyuki Yoneda
- Medicinal Chemistry Research Laboratory, Daiichi Pharmaceutical Co., Ltd., 16-13, Kitakasai 1-Chome, Edogawa-ku, 134-8630, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Harris NG, Gauden V, Fraser PA, Williams SR, Parker GJM. MRI measurement of blood-brain barrier permeability following spontaneous reperfusion in the starch microsphere model of ischemia. Magn Reson Imaging 2002; 20:221-30. [PMID: 12117604 DOI: 10.1016/s0730-725x(02)00498-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Quantification of the acute increases in blood-brain barrier (BBB) permeability that occur subsequent to experimental ischemic injury has been limited to single time-point, invasive methodologies. Although permeability can be qualitatively assessed to visualise regional changes during sequential studies on the same animal using contrast-enhanced magnetic resonance imaging (MRI), quantitative information on the magnitude of change is required to compare barrier function during sequential studies on the same animal or between different animals. Recently, improvements in MRI tracer kinetic models and in MR hardware design mean that an estimate of permeability in vivo can now be obtained with acceptable accuracy and precision. We report here the use of such methods to study acute changes following spontaneous reperfusion in an animal model of ischemia. We have obtained estimates of BBB permeability following spontaneous reperfusion, subsequent to forebrain ischemia by unilateral carotid injection of starch microspheres in the rat. T2*-weighted and diffusion-trace imaging were used to monitor the initial reduction in CBF and the time-course of ischemia, respectively. Following reperfusion, an intraveneous bolus of dimeglumine gadopentetate (Gd-DTPA) and horseradish peroxidase (HRP) was given during a continuous acquisition of T1 maps with a 48 s temporal resolution. Permeability maps were constructed using a 4-compartment model; K(trans), the permeability-surface area product of the capillary walls was estimated to be 9.2 +/- 0.6 x 10(-4) min(-1) in the cortex. Visualisation of the regional extent of HRP extravasation on histological sections following termination of the experiment demonstrated very little correspondence to the region of Gd-DTPA leakage. Quantitative MRI assessment of BBB permeability following ischemia-reperfusion is consistent with published values obtained by invasive methods. Differences between Gd-DTPA-enhancement and HRP may reflect differences in the molecular size of the tracers.
Collapse
Affiliation(s)
- Neil G Harris
- Unit of Biophysics, Institute of Child Health, University of London, London, United Kingdom.
| | | | | | | | | |
Collapse
|
36
|
Yoneda Y, Kawajiri S, Sugimura M, Osanai K, Kito F, Ota E, Mimura T. Synthesis of diaminobutane derivatives as potent Ca(2+)-permeable AMPA receptor antagonists. Bioorg Med Chem Lett 2001; 11:2663-6. [PMID: 11551773 DOI: 10.1016/s0960-894x(01)00530-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We synthesized diaminobutane derivatives as potent Ca(2+)-permeable AMPA receptor antagonists with non-hypotensive activity. Compound 10c showed selective Ca(2+)-permeable AMPA receptor antagonist activity and neuroprotective effects in transient global ischemia models in gerbils.
Collapse
Affiliation(s)
- Y Yoneda
- Medicinal Chemistry Research Laboratory, Daiichi Pharmaceutical Co., Ltd., 16-13, Kitakasai 1-Chome, Edogawa-ku, Tokyo 134-8630, Japan.
| | | | | | | | | | | | | |
Collapse
|
37
|
Sinha J, Das N, Basu MK. Liposomal antioxidants in combating ischemia-reperfusion injury in rat brain. Biomed Pharmacother 2001; 55:264-71. [PMID: 11428552 DOI: 10.1016/s0753-3322(01)00060-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Liposome-encapsulated antioxidants have been tested in vivo to prevent oxidative attack during cerebral ischemia and reperfusion. Oxidative stress is a causal factor in the neuropathogenesis of ischemic-reperfusion injury. From the therapeutic point of view free chemical antioxidants were almost ineffective to protect cerebral tissues from those oxidative attacks. Thus an attempt has been made to prevent the oxidative damage due to the cerebral ischemic insult by the introduction of chemical antioxidants, ascorbic acid and alpha-tocopherol either encapsulated or intercalated in small unilamellar liposomes. The effectiveness of antioxidant-loaded liposomes was tested against an experimental in vivo rat model of global cerebral ischemia. Oxidative free radical attack on cerebral tissues by the ischemic insult and brief reperfusion was accounted for by the amount of diene production per unit of tissue protein. Diene production in ischemic reperfused rat brain increases almost twofold over that of the normal rats. Prevention of excess diene production has been attributed to rats when they were treated either with L-ascorbic acid-encapsulated liposomes or alpha-tocopherol intercalated liposomes 2 hours prior to the cerebral ischemic insult. Complete restriction of excess diene generation has also been achieved when a mixture of alpha-tocopherol and L-ascorbic acid-encapsulated liposomes were injected 3 hours before the ischemic infraction.
Collapse
Affiliation(s)
- J Sinha
- Biomembrane Division, Indian Institute of Chemical Biology, Calcutta, India
| | | | | |
Collapse
|
38
|
Kagitani F, Uchida S, Hotta H, Sato A. Effects of nicotine on blood flow and delayed neuronal death following intermittent transient ischemia in rat hippocampus. THE JAPANESE JOURNAL OF PHYSIOLOGY 2000; 50:585-95. [PMID: 11173554 DOI: 10.2170/jjphysiol.50.585] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A cholinergic neural vasodilative response in the cerebral cortex and hippocampus, independent of metabolic vasodilation, was recently demonstrated by activating the nicotinic acetylcholine receptors (nAChRs) via activation of cholinergic neurons originating in the nucleus basalis of Meynert and septal complex in the basal forebrain and projecting to the cortex and hippocampus (see reviews by Sato A and Sato Y: Neurosci Res 14: 242--274, 1992; Sato A and Sato Y: Alzheimer Dis Assoc Disord 9: 28--38, 1995). In the present study, we aimed to examine whether an increase in regional blood flow in the hippocampus (Hpc-BF) following stimulation of the nAChRs by i.v. injection of nicotine could improve the delayed death of the hippocampal neurons following transient ischemia in rats. Hpc-BF was measured by using a laser Doppler flowmeter. During intermittent (every 2 min) transient occlusion for a total of 6 min of bilateral carotid arteries besides permanent ligation of bilateral vertebral arteries, Hpc-BF decreased to about 16% of the preocclusion level, and 5 or 7 d later, after the occlusion, delayed neuronal death occurred in approximately 70% of the CA1 hippocampal neurons. Hpc-BF was increased dose-dependently by injection of nicotine (30--100 microg/kg, i.v.), independent of mean arterial pressure. Nicotine (30--100 microg/kg) administered 5 min before occlusion slightly but significantly attenuated the occlusion-induced decrease in Hpc-BF. The delayed death of the CA1 hippocampal neurons occurring after transient occlusion was attenuated by pretreatment with nicotine (30--100 microg/kg) to approximately 50% of the total neurons. The results indicate that nAChR stimulation-induced increases in Hpc-BF can protect against ischemia-induced delayed death of hippocampal neurons.
Collapse
Affiliation(s)
- F Kagitani
- Department of the Autonomic Nervous System, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | | | | | | |
Collapse
|
39
|
van Lookeren Campagne M, Thomas GR, Thibodeaux H, Palmer JT, Williams SP, Lowe DG, van Bruggen N. Secondary reduction in the apparent diffusion coefficient of water, increase in cerebral blood volume, and delayed neuronal death after middle cerebral artery occlusion and early reperfusion in the rat. J Cereb Blood Flow Metab 1999; 19:1354-64. [PMID: 10598940 DOI: 10.1097/00004647-199912000-00009] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
It has been reported recently that very delayed damage can occur as a result of focal cerebral ischemia induced by vascular occlusion of short duration. With use of diffusion-, T2-, and contrast-enhanced dynamic magnetic resonance imaging (MRI) techniques, the occlusion time dependence together with the temporal profile for this delayed response in a rat model of transient focal cortical ischemia have been established. The distal branch of the middle cerebral artery was occluded for 20, 30, 45, or 90 minutes. Twenty minutes of vascular occlusion with reperfusion exhibited no significant mean change in either the apparent diffusion coefficient of water (ADC) or the T2 relaxation time at 6, 24, 48, or 72 hours after reperfusion (P = 0.97 and 0.70, respectively). Ninety minutes of ischemia caused dramatic tissue injury at 6 hours, as indicated by an increase in T2 relaxation times to 135% of the contralateral values (P < 0.01). However, at intermediate periods of ischemia (30 to 45 minutes), complete reversal of the ADC was seen at 6 hours after reperfusion but was followed by a secondary decline over time, such that a 25% reduction in tissue ADC was seen at 24 as compared with 6 hours (P < 0.02). This secondary response was accompanied by an increase in cerebral blood volume (CBV), as shown by contrast-enhanced dynamic MRI (120% of contralateral values; P < 0.001), an increase in T2 relaxation time (132%; P < 0.01), together with clear morphological signs of cell death. By day 18, the mean volume of missing cortical tissue measured with high-resolution MRI in animals occluded for 30 and 45 minutes was 50% smaller than that in 90-minute occluded animals (P < 0.005). These data show that ultimate infarct size is reduced after early reperfusion and is occlusion time dependent. The early tissue recovery that is seen with intermediate occlusion times can be followed by cell death, which has a delayed onset and is accompanied by an increase in CBV.
Collapse
Affiliation(s)
- M van Lookeren Campagne
- Department of Cardiovascular Research, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Rao AM, Hatcher JF, Kindy MS, Dempsey RJ. Arachidonic acid and leukotriene C4: role in transient cerebral ischemia of gerbils. Neurochem Res 1999; 24:1225-32. [PMID: 10492517 DOI: 10.1023/a:1020916905312] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Accumulation of arachidonic acid (AA) is greatest in brain regions most sensitive to transient ischemia. Free AA released after ischemia is either: 1) reincorporated into the membrane phospholipids, or 2) oxidized during reperfusion by lipoxygenases and cyclooxygenases, producing leukotrienes (LT), prostaglandins, thromboxanes and oxygen radicals. AA, its metabolite LTC4 and lipid peroxides (generated during AA metabolism) have been implicated in the blood-brain barrier (BBB) dysfunction, edema and neuronal death after ischemia/reperfusion. This report describes the time course of AA release, LTC4 accumulation and association with the physiological outcome during transient cerebral ischemia of gerbils. Significant amount of AA was detected immediately after 10 min ischemia (0 min reperfusion) which returned to sham levels within 30 min reperfusion. A later release of AA occurred after 1 d. LTC4 levels were elevated at 0-6 h and 1 d after ischemia. Increased lipid peroxidation due to AA metabolism was observed between 2-6 h. BBB dysfunction occurred at 6 h. Significant edema developed at 1 and 2 d after ischemia and reached maximum at 3 d. Ischemia resulted in approximately 80% neuronal death in the CA1 hippocampal region. Pretreatment with a 5-lipoxygenase inhibitor, AA861 resulted in significant attenuation of LTC4 levels (Baskaya et al. 1996. J. Neurosurg. 85: 112-116) and CA1 neuronal death. Accumulation of AA and LTC4, together with highly reactive oxygen radicals and lipid peroxides, may alter membrane permeability, resulting in BBB dysfunction, edema and ultimately to neuronal death.
Collapse
Affiliation(s)
- A M Rao
- Department of Neurological Surgery, University of Wisconsin, Madison 53792-3232, USA.
| | | | | | | |
Collapse
|
41
|
Pell GS, Lythgoe MF, Thomas DL, Calamante F, King MD, Gadian DG, Ordidge RJ. Reperfusion in a gerbil model of forebrain ischemia using serial magnetic resonance FAIR perfusion imaging. Stroke 1999; 30:1263-70. [PMID: 10356110 DOI: 10.1161/01.str.30.6.1263] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Existing methods for the quantitative measurement of the changing cerebral blood flow (CBF) during reperfusion suffer from poor spatial or temporal resolution. The aim of this study was to implement a recently developed MRI technique for quantitative perfusion imaging in a gerbil model of reperfusion. Flow-sensitive alternating inversion recovery (FAIR) is a noninvasive procedure that uses blood water as an endogenous tracer. METHODS Bilateral forebrain ischemia of 4 minutes' duration was induced in gerbils (n=8). A modified version of FAIR with improved time efficiency was used to provide CBF maps with a time resolution of 2.8 minutes after recirculation had been initiated. Quantitative diffusion imaging was also performed at intervals during the reperfusion period. RESULTS On initiating recirculation after the transient period of ischemia, the FAIR measurements demonstrated either a symmetrical, bilateral pattern of flow impairment (n=4) or an immediate side-to-side difference that became apparent with respect to the cerebral hemispheres in the imaged slice (n=4). The flow in each hemisphere displayed a pattern of recovery close to the preocclusion level or, alternatively, returned to a lower level before displaying a delayed hypoperfusion and a subsequent slow recovery. The diffusion measurements during this latter response suggested the development of cell swelling during the reperfusion phase in the striatum. CONCLUSIONS The CBF during the reperfusion period was monitored with a high time resolution, noninvasive method. This study demonstrates the utility of MRI techniques in following blood flow changes and their pathophysiological consequences.
Collapse
Affiliation(s)
- G S Pell
- Department of Medical Physics and Bioengineering, University College London, UK.
| | | | | | | | | | | | | |
Collapse
|
42
|
Pell GS, Thomas DL, Lythgoe MF, Calamante F, Howseman AM, Gadian DG, Ordidge RJ. Implementation of quantitative FAIR perfusion imaging with a short repetition time in time-course studies. Magn Reson Med 1999; 41:829-40. [PMID: 10332861 DOI: 10.1002/(sici)1522-2594(199904)41:4<829::aid-mrm24>3.0.co;2-u] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Flow-sensitive alternating inversion recovery (FAIR) is a pulsed arterial spin labeling magnetic resonance imaging method for perfusion quantification. In its standard implementation for quantification with full longitudinal relaxation between acquisitions, its use in time-course investigations of rapidly changing flow values is limited. The time efficiency can be improved by decreasing the repetition time but quantification becomes problematic. This situation is further complicated if a whole-body radiofrequency transmit coil is not used since fresh blood spins will flow in from outside the coil. To alleviate these problems, the use of global pre-saturation is proposed. The resulting expression for the flow signal depends on the relationship between the imaging parameters and the coil inflow time and can be significantly simplified under certain combinations of these parameters. With this implementation of FAIR, quantitative flow maps of gerbil brains were obtained with a 3 minute time resolution in a study of the effects of reperfusion. The pre-occlusion flow measurements were in good agreement with values obtained by the standard FAIR implementation and by other techniques, but the low values following occlusion were underestimated due to the increased transit times.
Collapse
Affiliation(s)
- G S Pell
- Department of Medical Physics and Bioengineering, University College London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
43
|
Ueda H, Tagawa K, Furuya E, Matsumoto M, Yanagihara T. A combined analysis of regional energy metabolism and immunohistochemical ischemic damage in the gerbil brain. J Neurochem 1999; 72:1232-42. [PMID: 10037496 DOI: 10.1046/j.1471-4159.1999.0721232.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
By combining immunohistochemical technique with microassay methods, we analyzed regional energy metabolism in vulnerable and tolerant areas of gerbil brains during evolution of neuronal damage after bilateral common carotid artery occlusion for 10 min with subsequent reperfusion. Four animals were used for each reperfusion period. Based on the information from the immunohistochemical examination, we dissected out vulnerable and tolerant subregions of the hippocampus, cerebral cortex, and thalamus from freeze-dried 20-microm-thick sections, and measured the levels of creatine phosphate (P-Cr), adenine nucleotides, guanine nucleotides, and purine bodies by HPLC, and the levels of glucose, glycogen, and lactate by an enzyme-immobilized column method. There were no significant differences in the levels of metabolites between vulnerable and tolerant subregions of control brains. After reperfusion, both vulnerable and tolerant subregions recovered preischemic metabolic profiles by 2 days. Although the regional differences between vulnerable and tolerant subregions were minimal at each reperfusion period, there were delays in the recovery of P-Cr, ATP, and/or total adenine nucleotides in all vulnerable subregions. A decline of P-Cr, ATP, and GTP levels without change in %ATP, AMP, or purine bodies occurred after reperfusion for 3 days, coinciding with the development of immunohistochemical damage by the immunoreaction for microtubule-associated protein 1A. The results supported the notion that subtle but sustained impairment of energy metabolism caused by mitochondrial dysfunction in the early reperfusion period might trigger delayed neuronal death in vulnerable subregions.
Collapse
Affiliation(s)
- H Ueda
- Department of Neurology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, USA
| | | | | | | | | |
Collapse
|
44
|
Shiino A, Matsuda M, Handa J, Chance B. Poor recovery of mitochondrial redox state in CA1 after transient forebrain ischemia in gerbils. Stroke 1998; 29:2421-4; discussion 2425. [PMID: 9804657 DOI: 10.1161/01.str.29.11.2421] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Several investigations have detected evidence of apoptosis in delayed neuronal death, but controversy prevails regarding this point. Recent studies have implicated mitochondria in apoptotic events. To explore relationships between delayed neuronal death and dysfunction of the respiratory chain, we analyzed mitochondrial redox changes in the gerbil hippocampus. METHODS We assessed the mitochondrial redox state in gerbil hippocampus before, during, and at various time points after 5 minutes of forebrain ischemia. The redox state was examined with a low-temperature fluorometer. Fluorescence signals of flavoprotein and NADH were measured, and their fluorescence ratio was calculated as a mitochondrial redox ratio (MRR) equal to flavoprotein/(flavoprotein+NADH). RESULTS Ischemia increased NADH and decreased flavoprotein signals in all hippocampal areas, but reduction in MRR was greater in CA1 than in other areas of the hippocampus. Immediately after recirculation, MRR recovery was delayed in the CA1 and the dentate gyrus, and the reduction in MRR persisted in CA1. CONCLUSIONS These results suggest that during ischemia CA1 experiences more pronounced hypoxia (state V) than less vulnerable regions. Persistent MRR reduction in CA1 is attributed to dysfunction of the electron transport system, and this phenomenon may be importantly involved in apoptosis.
Collapse
Affiliation(s)
- A Shiino
- Department of Neurosurgery, Shiga University of Medical Science, Ohtsu, Shiga, Japan.
| | | | | | | |
Collapse
|
45
|
Ichiki H, Kuroiwa T, Taniguchi I, Okeda R. Delayed recovery of auditory cortical evoked potentials is correlated with cortical neuronal death after transient cerebral ischemia in awake gerbils. Brain Res 1998; 806:278-81. [PMID: 9739151 DOI: 10.1016/s0006-8993(98)00757-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early detection of irreversible neuronal change after transient cerebral ischemia is important so that adequate treatment can be initiated within the therapeutic window. We have examined the correlation between changes in middle-latency auditory evoked potentials (MAEPs) and histological changes in the auditory cortex of awake Mongolian gerbils subjected to 4 min or 12 min of transient cerebral ischemia. Post-ischemic MAEPs were characterized by the appearance of a markedly large negative and positive component at approximately 17-22 ms latency in both groups. Delay in the appearance of the high amplitude (maximal amplitude at 45 min after recirculation in the 12-min ischemia group) precedes the slowly developing death of neurons in the auditory cortex that results from transient cerebral ischemia.
Collapse
Affiliation(s)
- H Ichiki
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Yushima 1-5-45, Bunkyo-ku, Tokyo, 113, Japan
| | | | | | | |
Collapse
|
46
|
Inci S, Ozcan OE, Kilinç K. Time-level relationship for lipid peroxidation and the protective effect of alpha-tocopherol in experimental mild and severe brain injury. Neurosurgery 1998; 43:330-5; discussion 335-6. [PMID: 9696087 DOI: 10.1097/00006123-199808000-00095] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Oxygen free radical-mediated lipid peroxidation has been proposed to be one of the major mechanisms of secondary damage in traumatic brain injury. The first purpose of this study was to establish the time-level relationship for lipid peroxidation in injured brain tissue. The second purpose was to examine the protective effect of alpha-tocopherol against lipid peroxidation. METHODS For this study, 65 guinea pigs in five groups were studied. Five of the animals were identified as a control group, and the remaining 60 animals were divided equally into four groups (Groups A, B, C, and D). Mild injury (200 g x cm) (Groups A and C) and severe injury (1000 g x cm) (Groups B and D) were produced by the method of Feeney et al. Alpha-tocopherol (100 mg/kg) was administered intraperitoneally before brain injury in Groups C and D. Five animals from each group were killed immediately after trauma, five after 1 hour, and the remaining five animals after 36 hours. Lipid peroxidation in traumatized brain tissues was assessed using the thiobarbituric acid method. RESULTS In all groups with traumatic brain injuries, levels of malondialdehyde, a lipid peroxidation product, were higher than in the control group. The amount of lipid peroxidation was increased by the severity of the trauma. Alpha-tocopherol significantly suppressed the rise in lipid peroxide levels in traumatized brain tissues. CONCLUSION This study demonstrates that lipid peroxidation is increased by the severity of trauma and that alpha-tocopherol has a protective effect against oxygen free radical-mediated lipid peroxidation in mild and severe brain injury.
Collapse
Affiliation(s)
- S Inci
- Department of Neurosurgery, School of Medicine, University of Hacettepe, Ankara, Turkey
| | | | | |
Collapse
|
47
|
Yamashima T, Kohda Y, Tsuchiya K, Ueno T, Yamashita J, Yoshioka T, Kominami E. Inhibition of ischaemic hippocampal neuronal death in primates with cathepsin B inhibitor CA-074: a novel strategy for neuroprotection based on 'calpain-cathepsin hypothesis'. Eur J Neurosci 1998; 10:1723-33. [PMID: 9751144 DOI: 10.1046/j.1460-9568.1998.00184.x] [Citation(s) in RCA: 197] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although Cornu Ammonis (CA) 1 neurons of the hippocampus are known to be vulnerable to transient ischaemia, the mechanism of ischaemic neuronal death is still unknown, and there are very few strategies to prevent neuronal death at present. In a previous report we demonstrated micro-calpain activation at the disrupted lysosomal membrane of postischaemic CA1 neurons in the monkey undergoing a complete 20 min whole brain ischaemia. Using the same experimental paradigm, we observed that the enzyme activity of the lysosomal protease cathepsin B increased throughout the hippocampus on days 3-5 after the transient ischaemia. Furthermore, by immunocytochemistry cathepsin B showed presence of extralysosomal immunoreactivity with specific localization to the cytoplasm of CA1 neurons and the neuropil of the vulnerable CA1 sector. When a specific inhibitor of cathepsin B, the epoxysuccinyl peptide CA-074 (C18H29N3O6) was intravenously administered immediately after the ischaemic insult, approximately 67% of CA1 neurons were saved from delayed neuronal death on day 5 in eight monkeys undergoing 20 min brain ischaemia: the extent of inhibition was excellent in three of eight and good in five of eight monkeys. The surviving neurons rescued by blockade of lysosomal activity, showed mild central chromatolysis and were associated with the decreased immunoreactivity for cathepsin B. These observations indicate that calpain-induced cathepsin B release is crucial for the development of the ischaemic neuronal death, and that a specific inhibitor of cathepsin B is of potential therapeutic utility in ischaemic injuries to the human CNS.
Collapse
Affiliation(s)
- T Yamashima
- Department of Neurosurgery, Kanazawa University School of Medicine, Takaramachi, Japan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Li JY, Ueda H, Seiyama A, Nakano M, Matsumoto M, Yanagihara T. A near-infrared spectroscopic study of cerebral ischemia and ischemic tolerance in gerbils. Stroke 1997; 28:1451-6; discussion 1456-7. [PMID: 9227699 DOI: 10.1161/01.str.28.7.1451] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE To explore the physiological mechanism of ischemic tolerance, we studied intracerebral oxygenation states noninvasively using near-infrared spectroscopy after bilateral common carotid artery occlusion (BCO) in gerbils with and without ischemic pretreatment. METHODS Under ether anesthesia, gerbils with sham operation (S group, n = 8) and those with pretreatment consisting of BCO for 2 minutes, twice at 3 days and 2 days earlier (T group, n = 8), were again subjected to BCO for 5 minutes. Changes in oxyhemoglobin (HbO2), deoxyhemoglobin (Hb), and total hemoglobin (HbT) as well as reduction in cytochrome oxidase (cyt.aa3) were calculated from the absorbance changes of the light transmitted through the brain. Seven days after the ischemic study, immunohistochemical examination was performed with an antiserum against microtubule-associated proteins. RESULTS In both groups, the increase of Hb and decrease of HbO2 and HbT proceeded rapidly after BCO, and the maximal deoxygenation of hemoglobin occurred within 2.5 minutes. Reduction of cyt.aa3 also ensued rapidly and reached the maximal reduction within 3 minutes in both groups. In the T group, however, both deoxygenation of hemoglobin and reduction of cyt.aa3 progressed more slowly than in the S group. The time (seconds) necessary for a maximal change for cyt.aa3 was significantly longer in the T group (203.8 +/- 34.0 [mean +/- SD]; P < .01) than in the S group (68.0 +/- 14.7). The time necessary for a half-maximal change was also significantly longer in the T group than in the S group for both Hb (22.0 +/- 7.5 and 13.5 +/- 4.0, respectively; P < .05) and cyt.aa3 (23.9 +/- 5.7 and 11.6 +/- 4.3; P < .01). After recirculation for 7 days, all gerbils in the S group were found to have neuronal death in the hippocampus, while those in the T group did not. CONCLUSIONS The present study indicated that mild ischemic stress can induce improvement in oxygen metabolism during subsequent ischemia, which might be causally related to the phenomenon known as "ischemic tolerance," in which a protective effect toward ischemic/postischemic injury is induced by earlier mild ischemic pretreatment.
Collapse
Affiliation(s)
- J Y Li
- Department of Neurology, Osaka University Medical School, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Başkaya MK, Rao AM, Doğan A, Donaldson D, Dempsey RJ. The biphasic opening of the blood-brain barrier in the cortex and hippocampus after traumatic brain injury in rats. Neurosci Lett 1997; 226:33-6. [PMID: 9153635 DOI: 10.1016/s0304-3940(97)00239-5] [Citation(s) in RCA: 237] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study examined the time course of the blood-brain barrier (BBB) opening and correlated this with brain edema formation after a lateral controlled cortical impact (CCI) brain injury in rats. Quantitative measurement of Evans blue (EB) extravasation using fluorescence was employed at 2, 4, 6 h and 1, 2, 3, 4 and 7 days after injury. Brain edema was measured by specific gravity of the tissue at corresponding time points. Two prominent EB extravasations were observed at 4-6 h and 3-day after injury in the injury-site cortex and the ipsilateral hippocampus. Brain edema became progressively more severe over time and peaked at 24 h after injury and began to decline after day 3. These results suggest that there is a biphasic opening of the BBB after CCI brain injury and the second opening of the BBB does not contribute to a further increase in edema formation.
Collapse
Affiliation(s)
- M K Başkaya
- Department of Neurological Surgery, University of Wisconsin, Madison 53792, USA
| | | | | | | | | |
Collapse
|
50
|
Asanuma M, Asanuma SN, Gómez-Vargas M, Yamamoto M, Ogawa N. Ketoprofen, a non-steroidal anti-inflammatory drug prevents the late-onset reduction of muscarinic receptors in gerbil hippocampus after transient forebrain ischemia. Neurosci Lett 1997; 225:109-12. [PMID: 9147386 DOI: 10.1016/s0304-3940(97)00204-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Ischemia-induced hippocampal late-onset reduction of muscarinic acetylcholine receptors (LORMAR) begins as late as 7 days after transient forebrain ischemia in the gerbil, but it precedes to completion of neuronal death in the CA1 region. We previously reported that post-ischemic administration of cyclosporin A prevented LORMAR with suppression of astroglial and microglial activation. In the present study, we showed that the chronic post-ischemic administration of a non-steroidal anti-inflammatory drug, ketoprofen (5 mg/kg, subcutaneously, twice a day for 14 days) significantly reduced LORMAR both 14 days and 21 days after 5-min transient ischemia. This protective effect of ketoprofen against LORMAR suggests that the non-steroidal anti-inflammatory drugs is clinically efficacious in the treatment of LORMAR, a sequela of cerebral ischemia.
Collapse
Affiliation(s)
- M Asanuma
- Department of Neuroscience, Okayama University Medical School, Japan
| | | | | | | | | |
Collapse
|