1
|
Sun M, Zhang Y, Mao R, Chen Y, Liu P, Ye L, Xu S, Jia J, Shu S, Li H, Yin Y, Xia S, Chen Y, Xu Y. MeCP2 Lactylation Protects against Ischemic Brain Injury by Transcriptionally Regulating Neuronal Apoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415309. [PMID: 40271828 DOI: 10.1002/advs.202415309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/25/2025] [Indexed: 04/25/2025]
Abstract
Lactate plays diverse roles in brain pathophysiology, including ischemic stroke. Here, the role of lysine lactylation, an epigenetic modification of lactate, in cerebral ischemia is investigated. Using a mouse model of transient middle cerebral artery occlusion, increased brain lactate levels and global protein lactylation are observed. Proteomics analysis reveals significant lactylation of non-histone proteins in the ischemic penumbra. Lactylation of MeCP2, a transcriptional regulator, is identified as a protective mechanism against stroke-induced neuronal death. Inhibition of MeCP2 lactylation through chemical or genetic manipulation increases infarct volume and aggravates neurological deficits. Mechanistically, MeCP2 lactylation at K210/K249 represses the transcription of apoptosis-associated genes, including Pdcd4 and Pla2g6, thereby attenuating neuronal apoptosis. Additionally, HDAC3 and p300 are identified as key enzymes that regulate MeCP2 lactylation post-stroke. The findings suggest that MeCP2 lactylation offers a potential therapeutic target for alleviating neuronal damage and improving stroke outcomes.
Collapse
Affiliation(s)
- Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuxin Zhang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Rui Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Yan Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Pinyi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Lei Ye
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Siyi Xu
- Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, 210008, China
| | - Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, 210008, China
| | - Shu Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Huiya Li
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Yanping Yin
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Yanting Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
- Nanjing Neurology Clinical Medical Center, and Nanjing Gulou Hospital Brain Disease and Brain Science Center, Nanjing, 210008, China
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Nanjing, 210008, China
| |
Collapse
|
2
|
Heiser H, Kiessler F, Roggenbach A, Ibanez V, Wieckhorst M, Helmchen F, Gjorgjieva J, Wahl AS. Brain-wide microstrokes affect the stability of memory circuits in the hippocampus. Nat Commun 2025; 16:3462. [PMID: 40216776 PMCID: PMC11992252 DOI: 10.1038/s41467-025-58688-4] [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: 10/10/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Cognitive deficits affect over 70% of stroke survivors, yet the mechanisms by which multiple small ischemic events contribute to cognitive decline remain poorly understood. In this study, we employed chronic two-photon calcium imaging to longitudinally track the fate of individual neurons in the hippocampus of mice navigating a virtual reality environment, both before and after inducing brain-wide microstrokes. Our findings reveal that, under normal conditions, hippocampal neurons exhibit varying degrees of stability in their spatial memory coding. However, microstrokes disrupted this functional network architecture, leading to cognitive impairments. Notably, the preservation of stable coding place cells, along with the stability, precision, and persistence of the hippocampal network, was strongly predictive of cognitive outcomes. Mice with more synchronously active place cells near important locations demonstrated recovery from cognitive impairment. This study uncovers critical cellular responses and network alterations following brain injury, providing a foundation for novel therapeutic strategies preventing cognitive decline.
Collapse
Affiliation(s)
- Hendrik Heiser
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Filippo Kiessler
- School of Life Sciences, Technical University of Munich, Maximus-von-Imhof-Forum 3, 85354, Freising, Germany
| | - Adrian Roggenbach
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Victor Ibanez
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Martin Wieckhorst
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland
- University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland
| | - Julijana Gjorgjieva
- School of Life Sciences, Technical University of Munich, Maximus-von-Imhof-Forum 3, 85354, Freising, Germany
| | - Anna-Sophia Wahl
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- Neuroscience Center Zurich (ZNZ), University of Zurich, Zurich, Switzerland.
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377, Munich, Germany.
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336, Munich, Germany.
| |
Collapse
|
3
|
Bonato JM, de Mattos BA, Oliveira DV, Milani H, Prickaerts J, de Oliveira RMW. Blood-Brain Barrier Rescue by Roflumilast After Transient Global Cerebral Ischemia in Rats. Neurotox Res 2023; 41:311-323. [PMID: 36922461 DOI: 10.1007/s12640-023-00639-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
Phosphodiesterase 4 inhibitors (PDE4-I), which selectively increase cyclic adenosine monophosphate (cAMP) levels, have shown neuroprotective effects after several neurological injuries inducing blood-brain barrier (BBB) damage including local/focal cerebral ischemia. The present investigated whether roflumilast confers BBB neuroprotection in the hippocampus after transient global cerebral ischemia (TGCI) in rats. TGCI resulted in whole BBB disruption as measured by the increase of Evans blue (EB) and IgG extravasation, neurodegeneration, and downregulation of claudin-5 and endothelial nitric oxide synthase (eNOS) levels in the CA1 hippocampal subfield of ischemic rats. Roflumilast attenuated BBB disruption and restored the levels of eNOS in the CA1 hippocampal area. Moreover, roflumilast increased the levels of B2 cell lymphoma (BcL-2) and neuron-glial antigen-2 (NG2) in the CA1 subfield after global ischemia in rats. The protective effects of roflumilast against TGCI-induced BBB breakdown might involve preservation of BBB integrity, vascularization and angiogenesis, and myelin repair.
Collapse
Affiliation(s)
- Jéssica Mendes Bonato
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, CEP, 5790, 87020-900, Maringá, Paraná, Brazil
| | - Bianca Andretto de Mattos
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, CEP, 5790, 87020-900, Maringá, Paraná, Brazil
| | - Daniela Velasquez Oliveira
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, CEP, 5790, 87020-900, Maringá, Paraná, Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, CEP, 5790, 87020-900, Maringá, Paraná, Brazil
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Rúbia Maria Weffort de Oliveira
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, CEP, 5790, 87020-900, Maringá, Paraná, Brazil.
| |
Collapse
|
4
|
Habashy KJ, Ahmad F, Ibeh S, Mantash S, Kobeissy F, Issa H, Habis R, Tfaily A, Nabha S, Harati H, Reslan MA, Yehya Y, Barsa C, Shaito A, Zibara K, El-Yazbi AF, Kobeissy FH. Western and ketogenic diets in neurological disorders: can you tell the difference? Nutr Rev 2022; 80:1927-1941. [PMID: 35172003 DOI: 10.1093/nutrit/nuac008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
The prevalence of obesity tripled worldwide between 1975 and 2016, and it is projected that half of the US population will be overweight by 2030. The obesity pandemic is attributed, in part, to the increasing consumption of the high-fat, high-carbohydrate Western diet, which predisposes to the development of the metabolic syndrome and correlates with decreased cognitive performance. In contrast, the high-fat, low-carbohydrate ketogenic diet has potential therapeutic roles and has been used to manage intractable seizures since the early 1920s. The brain accounts for 25% of total body glucose metabolism and, as a result, is especially susceptible to changes in the types of nutrients consumed. Here, we discuss the principles of brain metabolism with a focus on the distinct effects of the Western and ketogenic diets on the progression of neurological diseases such as epilepsy, Parkinson's disease, Alzheimer's disease, and traumatic brain injury, highlighting the need to further explore the potential therapeutic effects of the ketogenic diet and the importance of standardizing dietary formulations to assure the reproducibility of clinical trials.
Collapse
Affiliation(s)
| | - Fatima Ahmad
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarah Mantash
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Fatima Kobeissy
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hawraa Issa
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ralph Habis
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali Tfaily
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Comprehensive Epilepsy Program, Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Yehya
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Abdullah Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, and College of Medicine, Qatar University, Doha, Qatar
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Alalamein International University, Alalamein, Egypt
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience, and Chemistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
5
|
Sidtis JJ. Cerebral Blood Flow Is Not a Direct Surrogate of Behavior: Performance Models Suggest a Role for Functional Meta-Networks. Front Neurosci 2022; 16:771594. [PMID: 35242005 PMCID: PMC8885809 DOI: 10.3389/fnins.2022.771594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundFunctional brain imaging has become the dominant approach to the study of brain-behavior relationships. Unfortunately, the behavior half of the equation has been relegated to second-class status when it is not ignored completely. Different approaches to connectivity, based on temporally correlated physiological events across the brain, have ascended in place of behavior. A performance-based analysis has been developed as a simple, basic approach to incorporating specific performance measures obtained during imaging into the analysis of the imaging data identifying clinically relevant regions.MethodsThis paper contrasts performance-based lateralized regional cerebral blood flow (CBF) predictors of speech rate during Positron Emission Tomography with the values of these regions and their opposite hemisphere homologs in which a performance-based model was not applied. Five studies were examined: two that utilized normal speakers, one that utilized ataxic speakers, and two that examined Parkinsonian speakers.ResultsIn each study, the predictors were lateralized but the blood flow values that contributed to the performance-based analysis were bilateral. The speech-rate predictor regions were consistent with clinical studies on the effects of focal brain damage.ConclusionsThis approach has identified a basic, reproducible blood flow network that has predicted speech rate in multiple normal and neurologic groups. While the predictors are lateralized consistent with lesion data, the blood flow values of these regions are neither lateralized nor distinguished from their opposite hemisphere homologs in their magnitudes. The consistent differences between regional blood flow values and their corresponding regression coefficients in predicting performance suggests the presence of functional meta-networks that orchestrate the contributions of specific brain regions in support of mental and behavioral functions.
Collapse
Affiliation(s)
- John J. Sidtis
- Brain and Behavior Laboratory, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, New York University Langone School of Medicine, New York, NY, United States
- *Correspondence: John J. Sidtis,
| |
Collapse
|
6
|
Abstract
The susceptibility of the brain to ischaemic injury dramatically limits its viability following interruptions in blood flow. However, data from studies of dissociated cells, tissue specimens, isolated organs and whole bodies have brought into question the temporal limits within which the brain is capable of tolerating prolonged circulatory arrest. This Review assesses cell type-specific mechanisms of global cerebral ischaemia, and examines the circumstances in which the brain exhibits heightened resilience to injury. We suggest strategies for expanding such discoveries to fuel translational research into novel cytoprotective therapies, and describe emerging technologies and experimental concepts. By doing so, we propose a new multimodal framework to investigate brain resuscitation following extended periods of circulatory arrest.
Collapse
|
7
|
Khan MM, Badruddeen, Mujahid M, Akhtar J, Khan MI, Ahmad U. An Overview of Stroke: Mechanism, In vivo Experimental Models Thereof, and Neuroprotective Agents. Curr Protein Pept Sci 2021; 21:860-877. [PMID: 32552641 DOI: 10.2174/1389203721666200617133903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/22/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Stroke is one of the causes of death and disability globally. Brain attack is because of the acute presentation of stroke, which highlights the requirement for decisive action to treat it. OBJECTIVE The mechanism and in-vivo experimental models of stroke with various neuroprotective agents are highlighted in this review. METHOD The damaging mechanisms may proceed by rapid, nonspecific cell lysis (necrosis) or by the active form of cell death (apoptosis or necroptosis), depending upon the duration and severity and of the ischemic insult. RESULTS Identification of injury mediators and pathways in a variety of experimental animal models of global cerebral ischemia has directed to explore the target-specific cytoprotective strategies, which are critical to clinical brain injury outcomes. CONCLUSION The injury mechanism, available encouraging medicaments thereof, and outcomes of natural and modern medicines for ischemia have been summarized. In spite of available therapeutic agents (thrombolytics, calcium channel blockers, NMDA receptor antagonists and antioxidants), there is a need for an ideal drug for strokes.
Collapse
Affiliation(s)
- Mohd Muazzam Khan
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Badruddeen
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Mohd Mujahid
- Department of Pharmacology, College of Pharmacy, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | | | - Usama Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
8
|
Exercise preconditioning ameliorates cognitive impairment and anxiety-like behavior via regulation of dopamine in ischemia rats. Physiol Behav 2021; 233:113353. [PMID: 33571546 DOI: 10.1016/j.physbeh.2021.113353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/31/2020] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
Cognitive impairment and anxiety are common health problems in acute ischemic stroke patients. Meanwhile, dopamine in the striatal brain region is significantly increased during the acute phase of cerebral ischemia. Besides, the studies shown that striatum and change of striatal dopamine are associated with learning and memory and anxiety. Further, physical exercise has been shown to improve neurocognitive and emotional function in animal models and patients with cerebral ischemia. However, the exact mechanism underlying this effect is unclear. The purpose of this research is to explore the effect of pre-ischemic voluntary wheel running on levels of striatal dopamine, cognition and anxiety in cerebral ischemia rats. METHODS 48 adult male Sprague-Dawley rats were enrolled in this study and divided randomly in following 6 groups: sham group (S group, n = 8), ischemia group (I group, n = 8), 1 week wheel running group (1R group), 4 weeks wheel running group (4R group), 1 week pre-ischemia wheel running group (1RI group, n = 8) and 4 weeks pre-ischemia wheel running group (4RI group, n = 8). After training, cerebral ischemia was induced by permanent bilateral common carotid artery ligation (2-VO) operation. Microdialysis was used to collect dialysates from the striatum immediately from 30 min before ischemia to 90 min after ischemia. High-performance liquid chromatography-electrochemical detection system (HPLC) was used to determine the content of dopamine in the dialysates. Passive avoidance and elevated plus maze test were used to test neurocognitive function 24 h after 2-VO cerebral ischemia. RESULTS As compare with the constant striatal dopamine level of S group, the striatal dopamine level in I group after ischemia showed a trend of rapid increasing and reached maximum value at the 20 min (P<0.001), then decreased gradually. The striatal dopamine level in 1RI and 4RI group showed the trend were similar to I group, but the increasing magnitude was attenuated. A comparison of the basal striatal dopamine level in 4 groups found that the basal dopamine level in 1RI and 4RI group were higher than S and I group (P<0.001). In passive avoidance task, the retention latency of I group was significantly shorter than S group (P<0.001), and the retention latency of the 1RI, 1R and 4R, 4RI group were longer than I group (P<0.001), there was no significant difference in S, 1RI, 1R, 4R and 4RI group (P>0.05). In elevated plus maze test, the time and entrance numbers of open arms in I group were significantly less than S group (P<0.05), but these indices were no significant difference in S, 1RI, 1R, 4RI and 4RI group. CONCLUSION According to our results, 1 or 4 weeks pre-ischemia wheel running can significantly increase the basal dopamine level, attenuate the increase of striatal dopamine induced by cerebral ischemia and improve neurocognitive function in ischemia rats.
Collapse
|
9
|
Bonato JM, Meyer E, de Mendonça PSB, Milani H, Prickaerts J, Weffort de Oliveira RM. Roflumilast protects against spatial memory impairments and exerts anti-inflammatory effects after transient global cerebral ischemia. Eur J Neurosci 2021; 53:1171-1188. [PMID: 33340424 DOI: 10.1111/ejn.15089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Phosphodiesterase 4 (PDE4) inhibitors have been shown to present beneficial effects in cerebral ischemic injury because of their ability to improve cognition and target different phases and mechanisms of cerebral ischemia, including apoptosis, neurogenesis, angiogenesis, and inflammation. The present study investigated whether repeated treatment with the PDE4 inhibitor roflumilast rescued memory loss and attenuated neuroinflammation in rats following transient global cerebral ischemia (TGCI). TGCI caused memory impairments, neuronal loss (reflected by Neuronal nuclei (NeuN) immunoreactivity), and compensatory neurogenesis (reflected by doublecortin (DCX) immunoreactivity) in the hippocampus. Also, increases in the protein expression of the phosphorylated response element-binding protein (pCREB) and inflammatory markers such as the glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (Iba-1), were detected in the hippocampus in TGCI rats. Repeated treatment with roflumilast (0.003 and 0.01 mg/kg) prevented spatial memory deficits without promoting hippocampal protection in ischemic animals. Roflumilast increased the levels of pCREB, arginase-1, interleukin (IL) 4, and IL-10 in the hippocampus 21 days after TGCI. These data suggest a protective effect of roflumilast against functional sequelae of cerebral ischemia, which might be related to its anti-inflammatory properties.
Collapse
Affiliation(s)
- Jéssica M Bonato
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | - Erika Meyer
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | | | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
10
|
Shimizu Y, Harashima A, Munesue S, Oishi M, Hattori T, Hori O, Kitao Y, Yamamoto H, Leerach N, Nakada M, Yamamoto Y, Hayashi Y. Neuroprotective Effects of Endogenous Secretory Receptor for Advanced Glycation End-products in Brain Ischemia. Aging Dis 2020; 11:547-558. [PMID: 32489701 PMCID: PMC7220285 DOI: 10.14336/ad.2019.0715] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022] Open
Abstract
The receptor for advanced glycation end-products (RAGE) is expressed on human brain endothelial cells (HBEC) and is implicated in neuronal cell death after ischemia. We report that endogenous secretory RAGE (esRAGE) is a splicing variant form of RAGE that functions as a decoy against ischemia-induced neuronal cell damage. This study demonstrated that esRAGE was associated with heparan sulphate proteoglycans on HBEC. The parabiotic experiments between human esRAGE overexpressing transgenic (Tg), RAGE knockout (KO), and wild-type (WT) mice revealed a significant neuronal cell damage in the CA1 region of the WT side of parabiotic WT→WT mice, but not of Tg→WT mice, 7 days after bilateral common carotid artery occlusion. Human esRAGE was detected around the CA1 neurons in the WT side of the parabiotic Tg→WT pair, but not in the KO side of the Tg→KO pair. To elucidate the dynamic transfer of esRAGE into the brain, we used the blood-brain barrier (BBB) system (PharmaCo-Cell) with or without RAGE knockdown in endothelial cells. A RAGE-dependent transfer of esRAGE was demonstrated from the vascular to the brain side. These findings suggested that esRAGE is associated with heparan sulphate proteoglycans and is transferred into the brain via BBB to exert its neuroprotective effects in ischemia.
Collapse
Affiliation(s)
- Yu Shimizu
- 1Department of Biochemistry and Molecular Vascular Biology.,2Department of Neurosurgery and
| | - Ai Harashima
- 1Department of Biochemistry and Molecular Vascular Biology
| | | | - Masahiro Oishi
- 1Department of Biochemistry and Molecular Vascular Biology.,2Department of Neurosurgery and
| | - Tsuyoshi Hattori
- 3Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan
| | - Osamu Hori
- 3Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan
| | - Yasuko Kitao
- 3Department of Neuroanatomy, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8641, Japan
| | - Hiroshi Yamamoto
- 1Department of Biochemistry and Molecular Vascular Biology.,4Komatsu University, Komatsu, Ishikawa 923-8511, Japan
| | | | | | | | - Yasuhiko Hayashi
- 2Department of Neurosurgery and.,5Department of Neurosurgery, Kanazawa Medical University, Uchinada 920-0293, Japan
| |
Collapse
|
11
|
Keilhoff G, Nguyen Thi TM, Esser T, Ebmeyer U. Relative Resilience of Cerebellar Purkinje Cells in a Cardiac Arrest/Resuscitation Rat Model. Neurocrit Care 2019; 32:775-789. [DOI: 10.1007/s12028-019-00799-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
Lin CH, Chen SH, Chang CP, Lin KC. Hypothalamic impairment underlying heat intolerance in pregnant mice. Mol Cell Endocrinol 2019; 492:110439. [PMID: 31071379 DOI: 10.1016/j.mce.2019.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/18/2022]
Abstract
Pregnant women are vulnerable to heat stroke reactions caused by high environmental temperatures. Heat intolerance is associated with hypothalamic impairment. Here, we aim to ascertain whether pregnancy causes heat intolerance by inducing hypothalamic impairment in mice. In the heated groups, mice were exposed to whole body heating (WBH; 41.2 °C for 1 h) in an environment-controlled chamber. Then, they were returned to normal room temperature (26 °C) immediately after WBH. In the hyperbaric oxygen therapy (HBO2T) groups, mice were exposed to 100% O2 at 2.0 atm absolute (ATA) for 4 h immediately post-WBH. Mice that survived after 4 h of WBH were considered survivors. Here, we show that when pregnant mice underwent non-HBO2T (21% O2 at 1.0 ATA for 4 h) after WBH, the survival rate was 4/20, and the core temperature at 4 h post-WBH was 31.2 ± 0.2 °C. Both the survival rate and core temperature of HBO2T pregnant mice (10/10 and 35.2 ± 0.3 °C, respectively) were significantly greater than those in non-HBO2T pregnant mice. Compared to non-HBO2T heated mice, the HBO2T heated mice exhibited lower neurological severity scores, reduced hypothalamic neuronal damage, fewer apoptotic cells, reduced multiorgan damage scores, and lower hypothalamic levels of proinflammatory cytokines and nitrogen and oxygen radical species. Compared to non-HBO2T heated mice, the HBO2T-treated heated mice had significantly higher hypothalamic-pituitary-adrenal axis activity (evidenced by higher serum levels of both adrenocorticotrophic hormone and corticosterone). In conclusion, pregnancy induces heat intolerance by inducing hypothalamic impairment in mice. Additionally, HBO2T protects against heat intolerance in pregnant mice by preserving hypothalamic integrity.
Collapse
Affiliation(s)
- Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | | | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan City, Taiwan.
| | - Kao-Chang Lin
- Department of Neurology, Chi Mei Medical Center, Tainan City, Taiwan.
| |
Collapse
|
13
|
The Effects of Sodium Dichloroacetate on Mitochondrial Dysfunction and Neuronal Death Following Hypoglycemia-Induced Injury. Cells 2019; 8:cells8050405. [PMID: 31052436 PMCID: PMC6562710 DOI: 10.3390/cells8050405] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/17/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022] Open
Abstract
Our previous studies demonstrated that some degree of neuronal death is caused by hypoglycemia, but a subsequent and more severe wave of neuronal cell death occurs due to glucose reperfusion, which results from the rapid restoration of low blood glucose levels. Mitochondrial dysfunction caused by hypoglycemia leads to increased levels of pyruvate dehydrogenase kinase (PDK) and suppresses the formation of ATP by inhibiting pyruvate dehydrogenase (PDH) activation, which can convert pyruvate into acetyl-coenzyme A (acetyl-CoA). Sodium dichloroacetate (DCA) is a PDK inhibitor and activates PDH, the gatekeeper of glucose oxidation. However, no studies about the effect of DCA on hypoglycemia have been published. In the present study, we hypothesized that DCA treatment could reduce neuronal death through improvement of glycolysis and prevention of reactive oxygen species production after hypoglycemia. To test this, we used an animal model of insulin-induced hypoglycemia and injected DCA (100 mg/kg, i.v., two days) following hypoglycemic insult. Histological evaluation was performed one week after hypoglycemia. DCA treatment reduced hypoglycemia-induced oxidative stress, microglial activation, blood–brain barrier disruption, and neuronal death compared to the vehicle-treated hypoglycemia group. Therefore, our findings suggest that DCA may have the therapeutic potential to reduce hippocampal neuronal death after hypoglycemia.
Collapse
|
14
|
Ali SO, Shahin NN, Safar MM, Rizk SM. Therapeutic potential of endothelial progenitor cells in a rat model of epilepsy: Role of autophagy. J Adv Res 2019; 18:101-112. [PMID: 30847250 PMCID: PMC6389652 DOI: 10.1016/j.jare.2019.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 11/29/2022] Open
Abstract
This is the first report showing EPCs therapeutic effects in PTZ-induced epilepsy. Intravenously administered EPCs homed into the epileptic rat hippocampus. EPCs amend the memory and locomotor activity deficits related to epilepsy. EPCs ameliorate epilepsy-associated alterations in neurotransmitters and autophagy. EPCs mitigate concomitant histological and vascular anomalies.
Epilepsy is one of the most well-known neurological conditions worldwide. One-third of adult epileptic patients do not respond to antiepileptic drugs or surgical treatment and therefore suffer from the resistant type of epilepsy. Stem cells have been given substantial consideration in the field of epilepsy therapeutics. The implication of pathologic vascular response in sustained seizures and the eminent role of endothelial progenitor cells (EPCs) in maintaining vascular integrity tempted us to investigate the potential therapeutic effects of EPCs in a pentylenetetrazole (PTZ)-induced rat model of epilepsy. Modulation of autophagy, a process that enables neurons to maintain an equilibrium of synthesis, degradation and subsequent reprocessing of cellular components, has been targeted. Intravenously administered EPCs homed into the hippocampus and amended the deficits in memory and locomotor activity. The cells mitigated neurological damage and the associated histopathological alterations and boosted the expression of brain-derived neurotrophic factor. EPCs corrected the perturbations in neurotransmitter activity and enhanced the expression of the downregulated autophagy proteins light chain protein-3 (LC-3), beclin-1, and autophagy-related gene-7 (ATG-7). Generally, these effects were comparable to those achieved by the reference antiepileptic drug, valproic acid. In conclusion, EPCs may confer therapeutic effects against epilepsy and its associated behavioural and biochemical abnormalities at least in part via the upregulation of autophagy. The study warrants further research in experimental and clinical settings to verify the prospect of using EPCs as a valid therapeutic strategy in patients with epilepsy.
Collapse
Affiliation(s)
- Shimaa O Ali
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| | - Nancy N Shahin
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| | - Marwa M Safar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt.,Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, Egypt
| | - Sherine M Rizk
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| |
Collapse
|
15
|
Buscemi L, Price M, Bezzi P, Hirt L. Spatio-temporal overview of neuroinflammation in an experimental mouse stroke model. Sci Rep 2019; 9:507. [PMID: 30679481 PMCID: PMC6345915 DOI: 10.1038/s41598-018-36598-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/21/2018] [Indexed: 12/18/2022] Open
Abstract
After ischemic stroke, in the lesion core as well as in the ischemic penumbra, evolution of tissue damage and repair is strongly affected by neuroinflammatory events that involve activation of local specialized glial cells, release of inflammatory mediators, recruiting of systemic cells and vascular remodelling. To take advantage of this intricate response in the quest to devise new protective therapeutic strategies we need a better understanding of the territorial and temporal interplay between stroke-triggered inflammatory and cell death-inducing processes in both parenchymal and vascular brain cells. Our goal is to describe structural rearrangements and functional modifications occurring in glial and vascular cells early after an acute ischemic stroke. Low and high scale mapping of the glial activation on brain sections of mice subjected to 30 minutes middle cerebral artery occlusion (MCAO) was correlated with that of the neuronal cell death, with markers for microvascular changes and with markers for pro-inflammatory (IL-1β) and reparative (TGFβ1) cytokines. Our results illustrate a time-course of the neuroinflammatory response starting at early time-points (1 h) and up to one week after MCAO injury in mice, with an accurate spatial distribution of the observed phenomena.
Collapse
Affiliation(s)
- Lara Buscemi
- Stroke Laboratory, Neurology Service, Department of Clinical Neurosciences, University Hospital Centre and University of Lausanne, CH-1011, Lausanne, Switzerland. .,Department of Fundamental Neurosciences, University of Lausanne, CH-1005, Lausanne, Switzerland.
| | - Melanie Price
- Stroke Laboratory, Neurology Service, Department of Clinical Neurosciences, University Hospital Centre and University of Lausanne, CH-1011, Lausanne, Switzerland.,Department of Fundamental Neurosciences, University of Lausanne, CH-1005, Lausanne, Switzerland
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005, Lausanne, Switzerland
| | - Lorenz Hirt
- Stroke Laboratory, Neurology Service, Department of Clinical Neurosciences, University Hospital Centre and University of Lausanne, CH-1011, Lausanne, Switzerland.,Department of Fundamental Neurosciences, University of Lausanne, CH-1005, Lausanne, Switzerland
| |
Collapse
|
16
|
Luo XQ, Li A, Yang X, Xiao X, Hu R, Wang TW, Dou XY, Yang DJ, Dong Z. Paeoniflorin exerts neuroprotective effects by modulating the M1/M2 subset polarization of microglia/macrophages in the hippocampal CA1 region of vascular dementia rats via cannabinoid receptor 2. Chin Med 2018; 13:14. [PMID: 29560022 PMCID: PMC5859430 DOI: 10.1186/s13020-018-0173-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
Background Cerebral hypoperfusion is a pivotal risk factor for vascular dementia (VD), for which effective therapy remains inadequate. Persistent inflammatory responses and excessive chemotaxis of microglia/macrophages in the brain may accelerate the progression of VD. Endocannabinoids are involved in neuronal protection against inflammation-induced neuronal injury. Cannabinoids acting at cannabinoid receptor 2 (CB2R) can decrease inflammation. Based on the identification of paeoniflorin (PF) as a CB2R agonist, we investigated the neuroprotective and microglia/macrophages M1 to M2 polarization promoting effects of PF in a permanent four-vessel occlusion rat model. Methods One week after surgery, PF was intraperitoneally administered at a dose of 40 mg/kg once a day for 28 successive days. The effects of PF on memory deficit were investigated by a Morris water maze test, and the effects of PF on hippocampal neuronal damage were evaluated by light microscope and electron microscope. The mRNA and protein expression levels of key molecules related to the M1/M2 polarization of microglia/macrophages were assessed by RT-qPCR and Western blotting, respectively. Results Administration of PF could significantly attenuate cerebral hypoperfusion-induced impairment of learning and memory and reduce the morphological and ultrastructural changes in the hippocampal CA1 region of rats. Moreover, PF promoted an M1 to M2 phenotype transition in microglia/macrophages in the hippocampus of rats. In addition to its inhibitory property against proinflammatory M1 mediator expression, such as IL-1β, IL-6, TNF-α and NO, PF dramatically up-regulated expression of anti-inflammatory cytokines IL-10 and TGF-β1. Importantly, CB2R antagonist AM630 abolished these beneficial effects produced by PF on learning, memory and hippocampus structure in rats, as well as the polarization of microglia/macrophages to the M2 phenotype. Additionally, PF treatment significantly inhibited cerebral hypoperfusion-induced mTOR/NF-κB proinflammatory pathway and enhanced PI3K/Akt anti-inflammatory pathway. Effects of PF on these signaling pathways were effectively attenuated when rats were co-treated with PF and AM630, indicating that the mTOR/NF-κB and PI3K/Akt signaling pathways were involved in the PF effects through CB2R activation. Conclusion These findings demonstrated PF exerts its neuroprotective effect and shifts the inflammatory milieu toward resolution by modulation of microglia/macrophage polarization via CB2R activation. Electronic supplementary material The online version of this article (10.1186/s13020-018-0173-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xian-Qin Luo
- 1Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, 400016 China
| | - Ao Li
- 2College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054 China
| | - Xue Yang
- 3Institute of Chinese Pharmacology and Toxicology, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065 China
| | - Xiao Xiao
- 2College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054 China
| | - Rong Hu
- Drug Review Section, China Chongqing Technical Center for Drug Evaluation and Certification, Chongqing, 400014 China
| | - Tian-Wen Wang
- 3Institute of Chinese Pharmacology and Toxicology, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065 China
| | - Xiao-Yun Dou
- 5Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Da-Jian Yang
- 3Institute of Chinese Pharmacology and Toxicology, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065 China
| | - Zhi Dong
- 1Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, 400016 China
| |
Collapse
|
17
|
Lindblom RPF, Molnar M, Israelsson C, Röjsäter B, Wiklund L, Lennmyr F. Hyperglycemia Alters Expression of Cerebral Metabolic Genes after Cardiac Arrest. J Stroke Cerebrovasc Dis 2018; 27:1200-1211. [PMID: 29306595 DOI: 10.1016/j.jstrokecerebrovasdis.2017.11.036] [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: 09/14/2017] [Accepted: 11/26/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Survivors of cardiac arrest often experience neurologic deficits. To date, treatment options are limited. Associated hyperglycemia is believed to further worsen the neurologic outcome. The aim with this study was to characterize expression pathways induced by hyperglycemia in conjunction with global brain ischemia. METHODS Pigs were randomized to high or normal glucose levels, as regulated by glucose and insulin infusions with target levels of 8.5-10 mM and 4-5.5 mM, respectively. The animals were subjected to 5-minute cardiac arrest followed by 8 minutes of cardiopulmonary resuscitation and direct-current shock to restore spontaneous circulation. Global expression profiling of the cortex using microarrays was performed in both groups. RESULTS A total of 102 genes differed in expression at P < .001 between the hyperglycemic and the normoglycemic pigs. Several of the most strongly differentially regulated genes were involved in transport and metabolism of glucose. Functional clustering using bioinformatics tools revealed enrichment of multiple biological processes, including membrane processes, ion transport, and glycoproteins. CONCLUSIONS Hyperglycemia during cardiac arrest leads to differential early gene expression compared with normoglycemia. The functional relevance of these expressional changes cannot be deduced from the current study; however, the identified candidates have been linked to neuroprotective mechanisms and constitute interesting targets for further studies.
Collapse
Affiliation(s)
- Rickard Per Fredrik Lindblom
- Department of Cardiothoracic Surgery and Anaesthesia, Uppsala University Hospital, Uppsala, Sweden; Department of Surgical Sciences, Section of Thoracic Surgery, Uppsala University, Uppsala, Sweden
| | - Maria Molnar
- Department of Surgical Sciences, Section of Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.
| | - Charlotte Israelsson
- Department of Neuroscience, Developmental Neuroscience, Uppsala University, Uppsala, Sweden
| | - Belinda Röjsäter
- Department of Medical Sciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Wiklund
- Department of Surgical Sciences, Section of Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Fredrik Lennmyr
- Department of Cardiothoracic Surgery and Anaesthesia, Uppsala University Hospital, Uppsala, Sweden; Department of Surgical Sciences, Section of Thoracic Surgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Piao L, Fang YH, Kubler MM, Donnino MW, Sharp WW. Enhanced pyruvate dehydrogenase activity improves cardiac outcomes in a murine model of cardiac arrest. PLoS One 2017; 12:e0185046. [PMID: 28934276 PMCID: PMC5608301 DOI: 10.1371/journal.pone.0185046] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/04/2017] [Indexed: 11/19/2022] Open
Abstract
Rationale Post-ischemic changes in cellular metabolism alter myocardial and neurological function. Pyruvate dehydrogenase (PDH), the limiting step in mitochondrial glucose oxidation, is inhibited by increased expression of PDH kinase (PDK) during ischemia/reperfusion injury. This results in decreased utilization of glucose to generate cellular ATP. Post-cardiac arrest (CA) hypothermia improves outcomes and alters metabolism, but its influence on PDH and PDK activity following CA are unknown. We hypothesized that therapeutic hypothermia (TH) following CA is associated with the inhibition of PDK activity and increased PDH activity. We further hypothesized that an inhibitor of PDK activity, dichloroacetate (DCA), would improve PDH activity and post-CA outcomes. Methods and results Anesthetized and ventilated adult female C57BL/6 wild-type mice underwent a 12-minute KCl-induced CA followed by cardiopulmonary resuscitation. Compared to normothermic (37°C) CA controls, administering TH (30°C) improved overall survival (72-hour survival rate: 62.5% vs. 28.6%, P<0.001), post-resuscitation myocardial function (ejection fraction: 50.9±3.1% vs. 27.2±2.0%, P<0.001; aorta systolic pressure: 132.7±7.3 vs. 72.3±3.0 mmHg, P<0.001), and neurological scores at 72-hour post CA (9.5±1.3 vs. 5.4±1.3, P<0.05). In both heart and brain, CA increased lactate concentrations (1.9-fold and 3.1-fold increase, respectively, P<0.01), decreased PDH enzyme activity (24% and 50% reduction, respectively, P<0.01), and increased PDK protein expressions (1.2-fold and 1.9-fold, respectively, P<0.01). In contrast, post-CA treatment with TH normalized lactate concentrations (P<0.01 and P<0.05) and PDK expressions (P<0.001 and P<0.05), while increasing PDH activity (P<0.01 and P<0.01) in both the heart and brain. Additionally, treatment with DCA (0.2 mg/g body weight) 30 min prior to CA improved both myocardial hemodynamics 2 hours post-CA (aortic systolic pressure: 123±3 vs. 96±4 mmHg, P<0.001) and 72-hour survival rates (50% vs. 19%, P<0.05) in normothermic animals. Conclusions Enhanced PDH activity in the setting of TH or DCA administration is associated with improved post-CA resuscitation outcomes. PDH is a promising therapeutic target for improving post-CA outcomes.
Collapse
Affiliation(s)
- Lin Piao
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Yong-Hu Fang
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Manfred M. Kubler
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Michael W. Donnino
- Departments of Emergency Medicine and Internal Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Willard W. Sharp
- Section of Emergency Medicine; Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
19
|
Spray S, Johansson SE, Radziwon-Balicka A, Haanes KA, Warfvinge K, Povlsen GK, Kelly PAT, Edvinsson L. Enhanced contractility of intraparenchymal arterioles after global cerebral ischaemia in rat - new insights into the development of delayed cerebral hypoperfusion. Acta Physiol (Oxf) 2017; 220:417-431. [PMID: 27864916 DOI: 10.1111/apha.12834] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/11/2016] [Accepted: 11/15/2016] [Indexed: 12/13/2022]
Abstract
AIM Delayed cerebral hypoperfusion is a secondary complication found in the days after transient global cerebral ischaemia that worsens the ischaemic damage inflicted by the initial transient episode of global cerebral ischaemia. A recent study demonstrated increased cerebral vasoconstriction in the large arteries on the brain surface (pial arteries) after global cerebral ischaemia. However, smaller arterioles inside the brain (parenchymal arterioles) are equally important in the regulation of cerebral blood flow and yet their pathophysiology after global cerebral ischaemia is largely unknown. Therefore, we investigated whether increased contractility occurs in the intraparenchymal arterioles. METHODS Global cerebral ischaemia was induced in male Wistar rats by bilateral common carotid occlusion for 15 min combined with hypovolaemia. Regional cerebral blood flow was determined by quantitative autoradiography. Intraparenchymal arterioles were isolated and pressurized, and concentration-response curves to endothelin-1 with and without the endothelin B receptor-selective antagonist BQ788 was generated. Endothelin B receptor expression was investigated by quantitative flow cytometry and immunohistochemistry. RESULTS We observed increased endothelin-1-mediated contractility of parenchymal arterioles correlating with reduced cerebral blood flow of the cortex, hippocampus and caudate nucleus 48 h after global cerebral ischaemia. The increased endothelin-1-mediated contractility was abolished by BQ788, and the vascular smooth muscle cell-specific expression of endothelin B receptors was significantly increased after global cerebral ischaemia. CONCLUSION Increased endothelin-1-mediated contractility and expression of endothelin B receptors in the intraparenchymal vasculature contributes to the development of delayed cerebral hypoperfusion after global cerebral ischaemia in combination with vascular changes of the pial vasculature.
Collapse
Affiliation(s)
- S. Spray
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - S. E. Johansson
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - A. Radziwon-Balicka
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - K. A. Haanes
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - K. Warfvinge
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
- Division of Experimental Vascular Research; Department of Clinical Sciences; Lund University Hospital; Lund Sweden
| | - G. K. Povlsen
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - P. A. T. Kelly
- Centre for Cognitive and Neural System; University of Edinburgh; Edinburgh UK
| | - L. Edvinsson
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
- Division of Experimental Vascular Research; Department of Clinical Sciences; Lund University Hospital; Lund Sweden
| |
Collapse
|
20
|
Rehni AK, Liu A, Perez-Pinzon MA, Dave KR. Diabetic aggravation of stroke and animal models. Exp Neurol 2017; 292:63-79. [PMID: 28274862 PMCID: PMC5400679 DOI: 10.1016/j.expneurol.2017.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/03/2017] [Accepted: 03/03/2017] [Indexed: 12/16/2022]
Abstract
Cerebral ischemia in diabetics results in severe brain damage. Different animal models of cerebral ischemia have been used to study the aggravation of ischemic brain damage in the diabetic condition. Since different disease conditions such as diabetes differently affect outcome following cerebral ischemia, the Stroke Therapy Academic Industry Roundtable (STAIR) guidelines recommends use of diseased animals for evaluating neuroprotective therapies targeted to reduce cerebral ischemic damage. The goal of this review is to discuss the technicalities and pros/cons of various animal models of cerebral ischemia currently being employed to study diabetes-related ischemic brain damage. The rational use of such animal systems in studying the disease condition may better help evaluate novel therapeutic approaches for diabetes related exacerbation of ischemic brain damage.
Collapse
Affiliation(s)
- Ashish K Rehni
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Allen Liu
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
21
|
Dang YX, Shi KN, Wang XM. Early Changes in Glutamate Metabolism and Perfusion in Basal Ganglia following Hypoxia-Ischemia in Neonatal Piglets: A Multi-Sequence 3.0T MR Study. Front Physiol 2017; 8:237. [PMID: 28487658 PMCID: PMC5404207 DOI: 10.3389/fphys.2017.00237] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
The excitotoxicity of glutamate metabolism as well as hemodynamic disorders of the brain are both risk factors for neonatal hypoxic–ischemic brain damage (HIBD). In the present study, changes in glutamate metabolism in the basal ganglia were detected by proton magnetic resonance spectroscopy (1H-MRS) at 0–6, 8–12, 24–30, and 48–60 h after the induction of hypoxia-ischemia (HI) in newborn piglets. Meanwhile, correlation analysis was performed by combining the microcirculatory perfusion informations acquired by intravoxel incoherent motion (IVIM) scan to explore their possible interaction mechanism. The results suggested that Glu level in the basal ganglia underwent a “two-phase” change after HI; perfusion fraction f, an IVIM-derived perfusion parameter, was clearly decreased in the early stage after HI, then demonstrated a transient and slight recovery process, and thereafter continued to decrease. The changes in f and Glu level were in a significant negative correlation (r = −0.643, P = 0.001). Our study results revealed that Glu level is closely associated with the microcirculatory perfusion changes in the acute stage of HIBD.
Collapse
Affiliation(s)
- Yu-Xue Dang
- Department of Radiology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Kai-Ning Shi
- Department of Imaging Systems Clinical Science, Philips HealthcareBeijing, China
| | - Xiao-Ming Wang
- Department of Radiology, Shengjing Hospital of China Medical UniversityShenyang, China
| |
Collapse
|
22
|
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: 18] [Impact Index Per Article: 2.0] [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
|
23
|
Ikeda K, Liu X, Kida K, Marutani E, Hirai S, Sakaguchi M, Andersen LW, Bagchi A, Cocchi MN, Berg KM, Ichinose F, Donnino MW. Thiamine as a neuroprotective agent after cardiac arrest. Resuscitation 2016; 105:138-44. [PMID: 27185216 DOI: 10.1016/j.resuscitation.2016.04.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/19/2016] [Accepted: 04/25/2016] [Indexed: 11/19/2022]
Abstract
AIMS Reduction of pyruvate dehydrogenase (PDH) activity in the brain is associated with neurological deficits in animals resuscitated from cardiac arrest. Thiamine is an essential co-factor of PDH. The objective of this study was to examine whether administration of thiamine improves outcomes after cardiac arrest in mice. Secondarily, we aimed to characterize the impact of cardiac arrest on PDH activity in mice and humans. METHODS Animal study: Adult mice were subjected to cardiac arrest whereupon cardiopulmonary resuscitation was performed. Thiamine or vehicle was administered 2min before resuscitation and daily thereafter. Mortality, neurological outcome, and metabolic markers were evaluated. Human study: In a convenience sample of post-cardiac arrest patients, we measured serial PDH activity from peripheral blood mononuclear cells and compared them to healthy controls. RESULTS Animal study: Mice treated with thiamine had increased 10-day survival (48% versus 17%, P<0.01) and improved neurological function when compared to vehicle-treated mice. In addition, thiamine markedly improved histological brain injury compared to vehicle. The beneficial effects of thiamine were accompanied by improved oxygen consumption in mitochondria, restored thiamine pyrophosphate levels, and increased PDH activity in the brain at 10 days. Human study: Post-cardiac arrest patients had lower PDH activity in mononuclear cells than did healthy volunteers (estimated difference: -5.8O.D./min/mg protein, P<0.001). CONCLUSIONS The provision of thiamine after cardiac arrest improved neurological outcome and 10-day survival in mice. PDH activity was markedly depressed in post-cardiac arrest patients suggesting that this pathway may represent a therapeutic target.
Collapse
Affiliation(s)
- Kohei Ikeda
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Xiaowen Liu
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kotaro Kida
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Shuichi Hirai
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Masahiro Sakaguchi
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lars W Andersen
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Anaesthesiology, Aarhus University Hospital, Aarhus, Denmark; Research Center for Emergency Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Aranya Bagchi
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael N Cocchi
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Anesthesia Critical Care, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Katherine M Berg
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Division of Pulmonary and Critical Care, Beth Israel Deaconess Medical Center, MA, USA
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Michael W Donnino
- Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Division of Pulmonary and Critical Care, Beth Israel Deaconess Medical Center, MA, USA.
| |
Collapse
|
24
|
Rosafio K, Castillo X, Hirt L, Pellerin L. Cell-specific modulation of monocarboxylate transporter expression contributes to the metabolic reprograming taking place following cerebral ischemia. Neuroscience 2016; 317:108-20. [DOI: 10.1016/j.neuroscience.2015.12.052] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/17/2015] [Accepted: 12/29/2015] [Indexed: 01/23/2023]
|
25
|
Yeo LLL, Paliwal P, Low AF, Tay ELW, Gopinathan A, Nadarajah M, Ting E, Venketasubramanian N, Seet RCS, Ahmad A, Chan BPL, Teoh HL, Soon D, Rathakrishnan R, Sharma VK. How temporal evolution of intracranial collaterals in acute stroke affects clinical outcomes. Neurology 2016; 86:434-41. [PMID: 26740681 DOI: 10.1212/wnl.0000000000002331] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 10/09/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE We compared intracranial collaterals on pretreatment and day 2 brain CT angiograms (CTA) to assess their evolution and relationship with functional outcomes in acute ischemic stroke (AIS) patients treated with IV tissue plasminogen activator (tPA). METHODS Consecutive AIS patients who underwent pretreatment and day 2 CTA and received IV tPA during 2010-2013 were included. Collaterals were evaluated by 2 independent neuroradiologists using 3 predefined criteria: the Miteff system, the Maas system, and 20-point collateral scale by the Alberta Stroke Program Early CT Score methodology. We stratified our cohort by baseline pre-tPA state of their collaterals and by recanalization status of the primary vessel for analysis. Good outcomes at 3 months were defined by a modified Rankin Scale score of 0-1. RESULTS This study included 209 patients. Delayed collateral recruitment by any grading system was not associated with good outcomes. All 3 scoring systems showed that collateral recruitment on the follow-up CTA from a baseline poor collateral state was significantly associated with poor outcome and increased bleeding risk. When the primary vessel remained persistently occluded, collateral recruitment was significantly associated with worse outcomes. Interestingly, collateral recruitment was significantly associated with increased mortality in 2 of the 3 grading systems. CONCLUSIONS Not all collateral recruitment is beneficial; delayed collateral recruitment may be different from early recruitment and can result in worse outcomes and higher mortality. Prethrombolysis collateral status and recanalization are determinants of how intracranial collateral evolution affects functional outcomes.
Collapse
Affiliation(s)
- Leonard L L Yeo
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore.
| | - Prakash Paliwal
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Adrian F Low
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Edgar L W Tay
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Anil Gopinathan
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Mahendran Nadarajah
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Eric Ting
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Narayanaswamy Venketasubramanian
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Raymond C S Seet
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Aftab Ahmad
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Bernard P L Chan
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Hock L Teoh
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Derek Soon
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Rahul Rathakrishnan
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| | - Vijay K Sharma
- From the Division of Neurology, Department of Medicine (L.L.L.Y., P.P., R.C.S.S., A.A., B.P.L.C., H.L.T., D.S., R.R., V.K.S.), Cardiac Department (A.F.L., E.L.W.T.), and Department of Diagnostic Imaging (A.G., E.T.), National University Health System; Department of Neuroradiology (M.N.), National Neuroscience Institute; and Raffles Neuroscience Centre (N.V.), Raffles Medical Group, Singapore
| |
Collapse
|
26
|
|
27
|
Experimental animal models and inflammatory cellular changes in cerebral ischemic and hemorrhagic stroke. Neurosci Bull 2015; 31:717-34. [PMID: 26625873 DOI: 10.1007/s12264-015-1567-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/25/2015] [Indexed: 01/04/2023] Open
Abstract
Stroke, including cerebral ischemia, intracerebral hemorrhage, and subarachnoid hemorrhage, is the leading cause of long-term disability and death worldwide. Animal models have greatly contributed to our understanding of the risk factors and the pathophysiology of stroke, as well as the development of therapeutic strategies for its treatment. Further development and investigation of experimental models, however, are needed to elucidate the pathogenesis of stroke and to enhance and expand novel therapeutic targets. In this article, we provide an overview of the characteristics of commonly-used animal models of stroke and focus on the inflammatory responses to cerebral stroke, which may provide insights into a framework for developing effective therapies for stroke in humans.
Collapse
|
28
|
Varvarousis D, Varvarousi G, Iacovidou N, D'Aloja E, Gulati A, Xanthos T. The pathophysiologies of asphyxial vs dysrhythmic cardiac arrest: implications for resuscitation and post-event management. Am J Emerg Med 2015; 33:1297-304. [PMID: 26233618 DOI: 10.1016/j.ajem.2015.06.066] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/30/2015] [Accepted: 06/30/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Cardiac arrest is not a uniform condition and significant heterogeneity exists within all victims with regard to the cause of cardiac arrest. Primary cardiac (dysrhythmic) and asphyxial causes together are responsible for most cases of cardiac arrest at all age groups. The purpose of this article is to review the pathophysiologic differences between dysrhythmic and asphyxial cardiac arrest in the prearrest period, during the no-flow state, and after successful cardiopulmonary resuscitation. METHODS The electronic databases of PubMed/Medline, Scopus, and Cochrane were searched for relevant literature and studies. RESULTS/DISCUSSION Significant differences exist between dysrhythmic and asphyxial cardiac arrest regarding their pathophysiologic pathways and affect consequently the postresuscitation period. Laboratory data indicate that asphyxial cardiac arrest leads to more widespread postresuscitation brain damage compared with dysrhythmic cardiac arrest. Regarding postresuscitation myocardial dysfunction, few studies have addressed a comparison of the 2 conditions with controversial results. CONCLUSIONS Asphyxial cardiac arrest differs significantly from dysrhythmic cardiac arrest with regard to pathophysiologic mechanisms, neuropathologic damage, postresuscitation organ dysfunction, and response to therapy. Both conditions should be considered and treated in a different manner.
Collapse
Affiliation(s)
- Dimitrios Varvarousis
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Giolanda Varvarousi
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nicoletta Iacovidou
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ernesto D'Aloja
- Forensic Science Unit, Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, 09042 Monserrato, Italy
| | - Anil Gulati
- College of Pharmacy, Midwestern University, Downers Grove, IL
| | - Theodoros Xanthos
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; College of Pharmacy, Midwestern University, Downers Grove, IL
| |
Collapse
|
29
|
Pushkov D, Nicholson JD, Michowiz S, Novitzky I, Weiss S, Ben Hemou M, Hochhauser E, Goldenberg-Cohen N. Relative neuroprotective effects hyperbaric oxygen treatment and TLR4 knockout in a mouse model of temporary middle cerebral artery occlusion. Int J Neurosci 2015; 126:174-81. [PMID: 25562174 DOI: 10.3109/00207454.2014.1002609] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE To examine the effects of hyperbaric oxygen (HBO) therapy and knockout of toll-like receptor 4 (TLR4) on the outcome of temporary middle cerebral artery occlusion (MCAO) in a mouse model. MATERIALS AND METHODS MCAO was induced in anesthetized male C57Bl/6 mice (WT) and TLR4 knockout mice (TLR4(-/-)) using an intra-arterial filament method. After 30 or 90 min, the filament was removed, and the mice were given either no treatment (WT and TLR4(-/-) groups) or HBO (WT only). Mice were euthanized 24 h after MCAO, and the brain infarct area was examined using 2,3,5-triphenyltetrazolium chloride (TTC) staining. RESULTS In the WT group, without treatment, lesion volume was 120 ± 13 mm(3) in the mice subjected to 30 min' MCAO and 173 ± 23 mm(3) in the mice subjected to 90 min' MCAO. Respective values with HBO treatment were 66.5 ± 36.7 mm(3) and 53.2 ± 17.2 mm(3). The difference was significant only for 90-minute MCAO (p < 0.01, nonparametric test). In the TLR4(-/-) group (all untreated), lesion volume was 95.9 ± 17.9 after 90 min of MCAO, which was significantly lower than in the untreated WT animals (p < 0.05, nonparametric test). CONCLUSIONS A single treatment of HBO immediately after MCAO followed by 24 h' reperfusion significantly reduces edema and may improve perfusion. TLR4 knockout protects mice from MCAO damage, but to a lesser extent than HBO treatment.
Collapse
Affiliation(s)
- Dennis Pushkov
- a Department of Neurosurgery, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel
| | - James D Nicholson
- c Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,d The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Shalom Michowiz
- a Department of Neurosurgery, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel.,c Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ivan Novitzky
- a Department of Neurosurgery, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel
| | - Shirel Weiss
- c Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,d The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Moshe Ben Hemou
- c Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,d The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Edith Hochhauser
- c Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,e Laboratory of Cardiac Research, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Nitza Goldenberg-Cohen
- b Pediatric Ophthalmology Unit, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,c Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,d The Krieger Eye Research Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
30
|
Lapi D, Colantuoni A. Remodeling of Cerebral Microcirculation after Ischemia-Reperfusion. J Vasc Res 2015; 52:22-31. [DOI: 10.1159/000381096] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 02/15/2015] [Indexed: 11/19/2022] Open
|
31
|
Wang F, Shen Y, Tsuru E, Yamashita T, Baba N, Tsuda M, Maeda N, Sagara Y. Syngeneic transplantation of newborn splenocytes in a murine model of neonatal ischemia-reperfusion brain injury. J Matern Fetal Neonatal Med 2014; 28:842-7. [PMID: 24939627 DOI: 10.3109/14767058.2014.935327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Neonatal hypoxic-ischemic encephalopathy (HIE) is caused by brain injury that occurs in a developing fetus or infant. Stem cell transplantation can reportedly induce functional recovery in animal models of HIE. Murine neonatal splenocytes are enriched with immature blood stem cells and are used for the investigation of murine models of syngeneic transplantation. The aim of this study was to investigate the therapeutic potential of newborn splenocytes in a murine model of neonatal ischemia-reperfusion brain injury. METHODS C57BL/6N mice (postnatal day 7) underwent right common carotid artery occlusion with an aneurysm clip. Following hypoxic exposure, reperfusion was achieved by unclamping the artery. Newborn splenocytes were transplanted intravenously at 3 weeks after injury. RESULTS The splenocytes transplanted group tended to show an improvement in behavioral tests, but it was not significantly different compared with the control groups. The transplanted cells were localized in various organs including injured brain tissue over 3 weeks. In the penumbra region of the brain, vascular endothelial growth factor (VEGF) expression was upregulated after transplantation. CONCLUSIONS These results showed that syngeneic transplantation of newborn splenocytes achieved the long-term survival of the grafts and exerted influence the microenvironment in the injured brains of mice.
Collapse
Affiliation(s)
- Feifei Wang
- Center for Innovative and Translational Medicine, Kochi University Medical School , Nankoku, Kochi , Japan
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Katz LM, McGwin G, Gordon CJ. Drug-induced therapeutic hypothermia after asphyxial cardiac arrest in swine. Ther Hypothermia Temp Manag 2014; 2:176-82. [PMID: 24716490 DOI: 10.1089/ther.2012.0017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A feasibility study was performed to compare an investigational drug, HBN-1, to forced cooling to induce hypothermia after resuscitation in a translation model of asphyxial cardiac arrest in swine. Serum and cerebral spinal fluid neuron-specific enolase activity (sNSE and csfNSE) were measured after cardiac arrest as surrogate markers of brain injury. In a block design, swine resuscitated from 10 minutes of asphyxial cardiac arrest were infused intravenously with HBN-1 or iced saline vehicle (forced hypothermia [FH]) 5 to 45 minutes after return of spontaneous circulation (ROSC). External cooling in both groups was added 45 minutes after ROSC until hypothermia (T=4°C below baseline) was attained. Esophageal (core) temperature, shivering, cardiopulmonary parameters, and time to hypothermia after ROSC were monitored. sNSE and csfNSE were measured 180 minutes after ROSC. HBN-1 induced hypothermia significantly lowered temperature compared to FH 5-45 minutes after ROSC (p<0.0001). Time to hypothermia was reduced by HBN-1 (93±6 minutes) compared to FH (177±10 minutes) (p<0.0001). HBN-1 sNSE (0.7±1.9 ng/mL) and csfNSE (17.3±1.9 ng/mL) were lower compared to FH (6±1.6 ng/mL) and (49.7±32.0 ng/mL) (p<0.0001, p=0.022, respectively). There was no shivering with HBN-1 cooling while all FH cooled swine shivered (p<0.0001). The time to reach target hypothermia after cardiac arrest was reduced by nearly 50% with HBN-1 compared to the FH method of inducing hypothermia. Moreover, surrogate biomarkers of brain injury were significantly reduced with HBN-1 as compared to FH. While HBN-1-induced hypothermia shows promise for being neuroprotective, survival studies are needed to confirm these preliminary findings.
Collapse
Affiliation(s)
- Laurence M Katz
- 1 Department Emergency Medicine, University of North Carolina School of Medicine , Chapel Hill, North Carolina
| | | | | |
Collapse
|
33
|
Ultrasound-Microbubble Transplantation of Bone Marrow Stromal Cells Improves Neurological Function after Forebrain Ischemia in Adult Mice. Cell Biochem Biophys 2014; 70:499-504. [DOI: 10.1007/s12013-014-9947-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
34
|
Umbilical cord blood-derived stem cells improve heat tolerance and hypothalamic damage in heat stressed mice. BIOMED RESEARCH INTERNATIONAL 2014; 2014:685683. [PMID: 24804231 PMCID: PMC3997876 DOI: 10.1155/2014/685683] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 02/07/2014] [Accepted: 03/18/2014] [Indexed: 11/17/2022]
Abstract
Heatstroke is characterized by excessive hyperthermia associated with systemic inflammatory responses, which leads to multiple organ failure, in which brain disorders predominate. This definition can be almost fulfilled by a mouse model of heatstroke used in the present study. Unanesthetized mice were exposed to whole body heating (41.2°C for 1 hour) and then returned to room temperature (26°C) for recovery. Immediately after termination of whole body heating, heated mice displayed excessive hyperthermia (body core temperature ~42.5°C). Four hours after termination of heat stress, heated mice displayed (i) systemic inflammation; (ii) ischemic, hypoxic, and oxidative damage to the hypothalamus; (iii) hypothalamo-pituitary-adrenocortical axis impairment (reflected by plasma levels of both adrenocorticotrophic-hormone and corticosterone); (iv) decreased fractional survival; and (v) thermoregulatory deficits (e.g., they became hypothermia when they were exposed to room temperature). These heatstroke reactions can be significantly attenuated by human umbilical cord blood-derived CD34(+) cells therapy. Our data suggest that human umbilical cord blood-derived stem cells therapy may improve outcomes of heatstroke in mice by reducing systemic inflammation as well as hypothalamo-pituitary-adrenocortical axis impairment.
Collapse
|
35
|
Tseng LS, Chen SH, Lin MT, Lin YC. Transplantation of human dental pulp-derived stem cells protects against heatstroke in mice. Cell Transplant 2014; 24:921-37. [PMID: 24612725 DOI: 10.3727/096368914x678580] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stem cells from human exfoliated deciduous tooth pulp (SHED) is a promising approach for the treatment of stroke and spinal cord injury. In this study, we investigated the therapeutic effects of SHED for the treatment of multiple organ (including brain, particularly hypothalamus) injury in heatstroke mice. ICR male mice were exposed to whole body heating (WBH; 41.2°C, relative humidity 50-55%, for 1 h) and then returned to normal room temperature (26°C). We observed that intravenous administration of SHED immediately post-WBH exhibited the following therapeutic benefits for recovery after heatstroke: (a) inhibition of WBH-induced neurologic and thermoregulatory deficits; (b) reduction of WBH-induced ischemia, hypoxia, and oxidative damage to the brain (particularly the hypothalamus); (c) attenuation of WBH-induced increased plasma levels of systemic inflammatory response molecules, such as tumor necrosis factor-α and intercellular adhesion molecule-1; (d) improvement of WBH-induced hypothalamo-pituitary-adrenocortical (HPA) axis activity (as reflected by enhanced plasma levels of both adrenocorticotrophic hormone and corticosterone); and (e) attenuation of WBH-induced multiple organ apoptosis as well as lethality. In conclusion, post-WBH treatment with SHED reduced induction of proinflammatory cytokines and oxidative radicals, enhanced plasma induction of both adrenocorticotrophic hormone and corticosterone, and improved lethality in mouse heatstroke. The protective effect of SHED may be related to a decreased inflammatory response, decreased oxidative stress, and an increased HPA axis activity following the WBH injury.
Collapse
Affiliation(s)
- Ling-Shu Tseng
- School of Dentistry, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | | | | | | |
Collapse
|
36
|
Forebrain ischemia produces hippocampal damage and a persistent working memory deficit in rats. ACTA ACUST UNITED AC 2013. [DOI: 10.3758/bf03337687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Caldeira MV, Salazar IL, Curcio M, Canzoniero LMT, Duarte CB. Role of the ubiquitin-proteasome system in brain ischemia: friend or foe? Prog Neurobiol 2013; 112:50-69. [PMID: 24157661 DOI: 10.1016/j.pneurobio.2013.10.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/08/2013] [Accepted: 10/15/2013] [Indexed: 11/26/2022]
Abstract
The ubiquitin-proteasome system (UPS) is a catalytic machinery that targets numerous cellular proteins for degradation, thus being essential to control a wide range of basic cellular processes and cell survival. Degradation of intracellular proteins via the UPS is a tightly regulated process initiated by tagging a target protein with a specific ubiquitin chain. Neurons are particularly vulnerable to any change in protein composition, and therefore the UPS is a key regulator of neuronal physiology. Alterations in UPS activity may induce pathological responses, ultimately leading to neuronal cell death. Brain ischemia triggers a complex series of biochemical and molecular mechanisms, such as an inflammatory response, an exacerbated production of misfolded and oxidized proteins, due to oxidative stress, and the breakdown of cellular integrity mainly mediated by excitotoxic glutamatergic signaling. Brain ischemia also damages protein degradation pathways which, together with the overproduction of damaged proteins and consequent upregulation of ubiquitin-conjugated proteins, contribute to the accumulation of ubiquitin-containing proteinaceous deposits. Despite recent advances, the factors leading to deposition of such aggregates after cerebral ischemic injury remain poorly understood. This review discusses the current knowledge on the role of the UPS in brain function and the molecular mechanisms contributing to UPS dysfunction in brain ischemia with consequent accumulation of ubiquitin-containing proteins. Chemical inhibitors of the proteasome and small molecule inhibitors of deubiquitinating enzymes, which promote the degradation of proteins by the proteasome, were both shown to provide neuroprotection in brain ischemia, and this apparent contradiction is also discussed in this review.
Collapse
Affiliation(s)
- Margarida V Caldeira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Ivan L Salazar
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Portugal
| | - Michele Curcio
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Department of Science and Technology, University of Sannio, Benevento, Italy
| | | | - Carlos B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3004-517 Coimbra, Portugal.
| |
Collapse
|
38
|
Upregulated expression of NF-YC contributes to neuronal apoptosis via proapoptotic protein bim in rats' brain hippocampus following middle cerebral artery occlusion (MCAO). J Mol Neurosci 2013; 52:552-65. [PMID: 24014123 DOI: 10.1007/s12031-013-0111-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/27/2013] [Indexed: 01/10/2023]
Abstract
Cerebral ischemia represents a severe brain injury that could lead to significant neuronal damage and death. In this study, we performed a middle cerebral artery occlusion (MCAO) in adult rats and observed that a subunit of nuclear factor-Y (NF-Y) transcriptional factor, NF-YC, was accumulated in rat hippocampal CA1 neurons. Immunochemistrical and immunofluorescent analysis revealed that NF-YC was primarily expressed in the nucleus of neurons. Meanwhile, we found that the changes of bim, one of the target genes of NF-Y, were consistent with the expression of NF-YC and Bim was mainly located in the NF-YC positive cells. Moreover, there was a concomitant upregulation of active caspase-3 and TUNEL positive cells. Taken together, these results suggested that the upregulation of NF-YC might play an important role in the pathophysiology via proapoptotic protein Bim after MCAO and further research is needed to have a better understanding of its function and mechanism.
Collapse
|
39
|
Klopfleisch R. Multiparametric and semiquantitative scoring systems for the evaluation of mouse model histopathology--a systematic review. BMC Vet Res 2013; 9:123. [PMID: 23800279 PMCID: PMC3693904 DOI: 10.1186/1746-6148-9-123] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 06/19/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Histopathology has initially been and is still used to diagnose infectious, degenerative or neoplastic diseases in humans or animals. In addition to qualitative diagnoses semiquantitative scoring of a lesion`s magnitude on an ordinal scale is a commonly demanded task for histopathologists. Multiparametric, semiquantitative scoring systems for mouse models histopathology are a common approach to handle these questions and to include histopathologic information in biomedical research. RESULTS Inclusion criteria for scoring systems were a first description of a multiparametric, semiquantiative scoring systems which comprehensibly describe an approach to evaluate morphologic lesion. A comprehensive literature search using these criteria identified 153 originally designed semiquantitative scoring systems for the analysis of morphologic changes in mouse models covering almost all organs systems and a wide variety of disease models. Of these, colitis, experimental autoimmune encephalitis, lupus nephritis and collagen induced osteoarthritis colitis were the disease models with the largest number of different scoring systems. Closer analysis of the identified scoring systems revealed a lack of a rationale for the selection of the scoring parameters or a correlation between scoring parameter value and the magnitude of the clinical symptoms in most studies. CONCLUSION Although a decision for a particular scoring system is clearly dependent on the respective scientific question this review gives an overview on currently available systems and may therefore allow for a better choice for the respective project.
Collapse
Affiliation(s)
- Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
40
|
Arcangeli S, Tozzi A, Tantucci M, Spaccatini C, de Iure A, Costa C, Di Filippo M, Picconi B, Giampà C, Fusco FR, Amoroso S, Calabresi P. Ischemic-LTP in striatal spiny neurons of both direct and indirect pathway requires the activation of D1-like receptors and NO/soluble guanylate cyclase/cGMP transmission. J Cereb Blood Flow Metab 2013; 33:278-86. [PMID: 23149555 PMCID: PMC3564198 DOI: 10.1038/jcbfm.2012.167] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Striatal medium-sized spiny neurons (MSNs) are highly vulnerable to ischemia. A brief ischemic insult, produced by oxygen and glucose deprivation (OGD), can induce ischemic long-term potentiation (i-LTP) of corticostriatal excitatory postsynaptic response. Since nitric oxide (NO) is involved in the pathophysiology of brain ischemia and the dopamine D1/D5-receptors (D1-like-R) are expressed in striatal NOS-positive interneurons, we hypothesized a relation between NOS-positive interneurons and striatal i-LTP, involving D1R activation and NO production. We investigated the mechanisms involved in i-LTP induced by OGD in corticostriatal slices and found that the D1-like-R antagonist SCH-23390 prevented i-LTP in all recorded MSNs. Immunofluorescence analysis confirmed the induction of i-LTP in both substance P-positive, (putative D1R-expressing) and adenosine A2A-receptor-positive (putative D2R-expressing) MSNs. Furthermore, i-LTP was dependent on a NOS/cGMP pathway since pharmacological blockade of NOS, guanylate-cyclase, or PKG prevented i-LTP. However, these compounds failed to prevent i-LTP in the presence of a NO donor or cGMP analog, respectively. Interestingly, the D1-like-R antagonism failed to prevent i-LTP when intracellular cGMP was pharmacologically increased. We propose that NO, produced by striatal NOS-positive interneurons via the stimulation of D1-like-R located on these cells, is critical for i-LTP induction in the entire population of MSNs involving a cGMP-dependent pathway.
Collapse
Affiliation(s)
- Sara Arcangeli
- Clinica Neurologica, Università di Perugia, Ospedale S. Maria della Misericordia, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhai D, Li S, Wang M, Chin K, Liu F. Disruption of the GluR2/GAPDH complex protects against ischemia-induced neuronal damage. Neurobiol Dis 2013; 54:392-403. [PMID: 23360709 DOI: 10.1016/j.nbd.2013.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 12/29/2012] [Accepted: 01/17/2013] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Excitotoxicity and neuronal death following ischemia involve AMPA (α-amino-3hydroxy-5-methylisoxazole-4-propionic acid) glutamate receptors. We have recently reported that the GluR2 subunit of AMPA receptors (AMPARs) forms a protein complex with GAPDH (glyceraldehyde-3-phosphate dehydrogenase). The GluR2/GAPDH complex co-internalizes upon activation of AMPA receptors. Disruption of the GluR2/GAPDH interaction with an interfering peptide protects cells against AMPAR-mediated excitotoxicity and protects against damage induced by oxygen-glucose deprivation (OGD), an in vitro model of brain ischemia. OBJECTIVE We sought to test the hypothesis that disruption of the GluR2/GAPDH interaction with an interfering peptide would protect against ischemia-induced neuronal damage in vivo. METHOD The rat 4-vessel occlusion (4-VO) model was used to investigate whether the GluR2/GAPDH interaction was enhanced in the hippocampus, and if our newly developed interfering peptide could protect against neuronal death in the ischemic brain area. The transient rat middle cerebral artery occlusion (tMCAo) model was used to determine whether our peptide could reduce infarction volume and improve neurological function. Finally, GAPDH lentiviral shRNA was injected into the brain to reduce GAPDH expression one week prior to tMCAo, to confirm the role of GAPDH in the pathophysiology of brain ischemia. RESULTS The GluR2/GAPDH interaction is upregulated in the hippocampus of rats subjected to transient global ischemia. Administration of an interfering peptide that is able to disrupt the GluR2/GAPDH interaction in vivo protects against ischemia-induced cell death in rat models of global ischemia and decreases the infarct volume as well as neurological score in a rat model focal ischemia. Consistent with these observations, decreased GAPDH expression also protects against ischemia-induced cell death in an animal model of focal ischemia. CONCLUSION Disruption of the GluR2/GAPDH interaction protects against ischemia-induced neuronal damage in vivo. The GluR2/GAPDH interaction may be a novel therapeutic target for development of treatment for ischemic stroke.
Collapse
Affiliation(s)
- Dongxu Zhai
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8 Canada
| | | | | | | | | |
Collapse
|
42
|
Jung YJ, Suh EC, Lee KE. Oxygen/Glucose Deprivation and Reperfusion Cause Modifications of Postsynaptic Morphology and Activity in the CA3 Area of Organotypic Hippocampal Slice Cultures. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:423-9. [PMID: 23269905 PMCID: PMC3526747 DOI: 10.4196/kjpp.2012.16.6.423] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/08/2012] [Accepted: 10/20/2012] [Indexed: 12/25/2022]
Abstract
Brain ischemia leads to overstimulation of N-methyl-D-aspartate (NMDA) receptors, referred as excitotoxicity, which mediates neuronal cell death. However, less attention has been paid to changes in synaptic activity and morphology that could have an important impact on cell function and survival following ischemic insult. In this study, we investigated the effects of reperfusion after oxygen/glucose deprivation (OGD) not only upon neuronal cell death, but also on ultrastructural and biochemical characteristics of postsynaptic density (PSD) protein, in the stratum lucidum of the CA3 area in organotypic hippocampal slice cultures. After OGD/reperfusion, neurons were found to be damaged; the organelles such as mitochondria, endoplasmic reticulum, dendrites, and synaptic terminals were swollen; and the PSD became thicker and irregular. Ethanolic phosphotungstic acid staining showed that the density of PSD was significantly decreased, and the thickness and length of the PSD were significantly increased in the OGD/reperfusion group compared to the control. The levels of PSD proteins, including PSD-95, NMDA receptor 1, NMDA receptor 2B, and calcium/calmodulin-dependent protein kinase II, were significantly decreased following OGD/reperfusion. These results suggest that OGD/reperfusion induces significant modifications to PSDs in the CA3 area of organotypic hippocampal slice cultures, both morphologically and biochemically, and this may contribute to neuronal cell death and synaptic dysfunction after OGD/reperfusion.
Collapse
Affiliation(s)
- Yeon Joo Jung
- Department of Pharmacology and Ewha Medical Research Institute, Ewha Womans University School of Medicine, Seoul 158-710, Korea
| | | | | |
Collapse
|
43
|
Abstract
Anoxic depolarization of pyramidal neurons results from a large inward current that is activated, in part, by excessive glutamate release during exposure to anoxia/ischemia. Pannexin-1 (Panx1) channels can be activated both by ischemia and NMDA receptors (NMDARs), but the mechanisms of Panx1 activation are unknown. We used whole-cell recordings to show that pharmacological inhibition or conditional genetic deletion of Panx1 strongly attenuates the anoxic depolarization of CA1 pyramidal neurons in acute brain slices from rats and mice. Anoxia or exogenous NMDA activated Src family kinases (SFKs), as measured by increased phosphorylation of SFKs at Y416. The SFK inhibitor PP2 prevented Src activation and Panx1 opening during anoxia. A newly developed interfering peptide that targets the SFK consensus-like sequence of Panx1 (Y308) attenuated the anoxic depolarization (AD) without affecting SFK activation. Importantly, the NMDAR antagonists, D-APV and R-CPP, attenuated AD currents carried by Panx1, and the combined application of D-APV and (10)panx (a Panx1 blocker) inhibited AD currents to the same extent as either blocker alone. We conclude that activation of NMDARs during anoxia/ischemia recruits SFKs to open Panx1, leading to sustained neuronal depolarizations.
Collapse
|
44
|
Gu Q, Gu H, Lu X, Lu F, Shao Y, Zhang S. The influence of deep hypothermic global brain ischemia on EEG in a new rat model. J Card Surg 2012; 27:612-7. [PMID: 22978840 DOI: 10.1111/j.1540-8191.2012.01498.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Neurological complications following deep hypothermic circulatory arrest (DHCA) occur between t 4% ≈ 25%. However, the cerebral injury mechanisms are still not well understood due to a lack of a practical and simple animal model. We aimed to establish a rodent deep hypothermic global brain ischemia (DHGBI) model, which can be used to elucidate these mechanisms in future studies. DESIGN 30 Sprague-Dawley rats were divided randomly into three groups: the carotid occlusion DHGBI group, the internal carotid shunt DHGBI group, and the sham operation group. We validated the model in terms of electroencephalogram (EEG) and regional cerebral blood flow (rCBF). All rats were sacrificed for analysis of brain moisture capacity after 24 hours. RESULTS In the internal carotid shunt DHGBI group the EEG activity was suppressed to "flat-line" and the relative power of the α and θ frequency bands was decreased (p < 0.05). However, in the carotid occlusion DHGBI group we only observed the relative power of the α frequency band depressed (p < 0.05). The rCBF was significantly decreased in all groups. In the internal carotid shunt DHGBI group the rCBF was significantly reduced to 4.27 ± 2.75%, and was lower than the other two groups (p < 0.05). The result of brain moisture capacity was consistent with the EEG and rCBF observations. CONCLUSIONS The current study presents a novel cerebral recovery model of DHCA in the rat. This experimental model may be suitable to further elucidate the mechanisms associated with adverse cerebral outcomes after DHCA and to investigate potential neuroprotective strategies.
Collapse
Affiliation(s)
- Qun Gu
- Jiangsu Province Hospital, First Affiliated Hospital with Nanjing Medical University, Department of Cardiothoracic Surgery, Nanjing, China
| | | | | | | | | | | |
Collapse
|
45
|
Attenuating brain inflammation, ischemia, and oxidative damage by hyperbaric oxygen in diabetic rats after heat stroke. J Formos Med Assoc 2012; 112:454-62. [PMID: 24016610 DOI: 10.1016/j.jfma.2012.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND/PURPOSE Alternating hypothalamic-pituitary-adrenal axis mechanisms would lead to multiple organs dysfunction or failure. Herein, we attempt to assess whether hypothalamic inflammation and ischemic and oxidative damage that occurred during heatstroke (HS) can be affected by hyperbaric oxygen (HBO₂) therapy in streptozotocin-induced diabetic rats. METHODS In this study, anesthetized diabetic rats, immediately after the onset of HS, were divided into two major groups and given the normobaric air (21% O₂ at 1.0 atmospheres absolute) or HBO₂ (100% O₂ at 2.0 atmospheres absolute). HS was induced by exposing the animals to heat stress (43°C). Another group of anesthetized diabetic rats was kept at normothermic state and used as controls. RESULTS The survival time values for the HBO2-treated HS-diabetic rats increased form the control values of 78-82 minutes to new values of 184-208 minutes. HBO₂ therapy caused a reduction of HS-induced cellular ischemia (e.g., increased cellular levels of glutamate and lactate/pyruvate ratio), hypoxia (e.g., decreased cellular levels of PO₂), inflammation (e.g., increased cellular levels of interleukin-1β, tumor necrosis factor-alpha, interleukin-6, and myeloperoxidase), and oxidative damage (e.g., increased values of nitric oxide, 2,3-dihydroxybenzoic acid, glycerol, and neuronal damage score) in the hypothalamus of the diabetic rats. CONCLUSION Our results suggest that, in diabetic animals, HBO2 therapy may improve outcomes of HS in part by reducing heat-induced activated inflammation and ischemic and oxidative damage in the hypothalamus and other brain regions.
Collapse
|
46
|
Gaynes B, Teng PY, Wanek J, Shahidi M. Feasibility of conjunctival hemodynamic measurements in rabbits: reproducibility, validity, and response to acute hypotension. Microcirculation 2012; 19:521-9. [PMID: 22486988 PMCID: PMC3648337 DOI: 10.1111/j.1549-8719.2012.00182.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To evaluate the feasibility of conjunctival hemodynamic measurements based on assessment of reproducibility, validity, and response to acute hypotension. METHODS Image sequences of the conjunctival microvasculature of rabbits were captured using a slit lamp biomicroscope under a steady-state condition, after topical administration of phenylephrine, and after intravenous administration of esmolol. Venous hemodynamic parameters (diameter, blood velocity, blood flow, and wall shear stress) were derived. RESULTS Conjunctival venous diameters ranged from 9 to 34 μm and blood velocities ranged from 0.08 to 0.95 mm/s. Coefficients of variation of venous diameter and blood velocity measurements were, on average, 6% and 14%, respectively. Automated and manual measurements of venous diameter and velocity were highly correlated (R = 0.97; p < 0.001; n = 16). With phenylephrine administration, diameter and velocity were reduced by 21% and 69%, respectively. Following esmolol administration, blood pressure was reduced with a concomitant decrease in velocity, followed by recovery to baseline. Venous blood velocity, flow, and WSS were correlated with blood pressure (R ≥ 0.52; p ≤ 0.01). CONCLUSIONS The feasibility of quantifying alterations in microvascular hemodynamics in the bulbar conjunctiva was established. The method is of potential value in evaluating microcirculatory hemodynamics related to cardiovascular function.
Collapse
Affiliation(s)
- Bruce Gaynes
- Department of Ophthalmology, Loyola University Medical Center, Maywood, Illinois, USA
| | | | | | | |
Collapse
|
47
|
Wang M, Li S, Zhang H, Pei L, Zou S, Lee FJS, Wang YT, Liu F. Direct interaction between GluR2 and GAPDH regulates AMPAR-mediated excitotoxicity. Mol Brain 2012; 5:13. [PMID: 22537872 PMCID: PMC3407747 DOI: 10.1186/1756-6606-5-13] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Accepted: 04/26/2012] [Indexed: 12/30/2022] Open
Abstract
Over-activation of AMPARs (α−amino-3-hydroxy-5-methylisoxazole-4-propionic acid subtype glutamate receptors) is implicated in excitotoxic neuronal death associated with acute brain insults, such as ischemic stroke. However, the specific molecular mechanism by which AMPARs, especially the calcium-impermeable AMPARs, induce neuronal death remains poorly understood. Here we report the identification of a previously unrecognized molecular pathway involving a direct protein-protein interaction that underlies GluR2-containing AMPAR-mediated excitotoxicity. Agonist stimulation of AMPARs promotes GluR2/GAPDH (glyceraldehyde-3-phosphate dehydrogenase) complex formation and subsequent internalization. Disruption of GluR2/GAPDH interaction by administration of an interfering peptide prevents AMPAR-mediated excitotoxicity and protects against damage induced by oxygen-glucose deprivation (OGD), an in vitro model of brain ischemia.
Collapse
Affiliation(s)
- Min Wang
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, Canada
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim YT, Yi YJ, Kim MY, Bu Y, Jin ZH, Choi H, Doré S, Kim H. Neuroprotection and Enhancement of Spatial Memory by Herbal Mixture HT008-1 in Rat Global Brain Ischemia Model. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 36:287-99. [DOI: 10.1142/s0192415x08005771] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To investigate whether HT008-1, a prescription used in traditional Korean medicine to treat mental and physical weakness, has a neuroprotective effect on a rat model of global brain ischemia and an enhancing effect against memory deficit following ischemia. Global brain ischemia was induced for 10 min by using 4-vessel occlusion (4-VO). HT008-1 was orally administered at doses of 30, 100, and 300 mg/kg respectively twice at 0 and 90 min after ischemia. The effect on memory deficit was investigated by using a Y-maze neurobehavioral test 4 days after brain ischemia, and the effect on neuronal damage was measured 7 days after ischemia. The mechanism of action was studied immunohistochemically using an anti-CD11b (OX-42) antibody. The oral administration of HT008-1 at 100 and 300 mg/kg significantly reduced hippocampal neuronal cell death by 49% and 53%, respectively, compared with a vehicle-treated group, and also improved spatial memory function in the Y-maze test. Immunohistochemically, HT008-1 inhibited OX-42 expression in the hippocampus. The effects of HT008-1 were more pronounced than those of its individual herb components. The herbal mixture HT008-1 protects the most vulnerable CA1 pyramidal cells of the hippocampus and enhances spatial memory function against global brain ischemia; an anti-inflammatory effect may be one of the mechanisms of action.
Collapse
Affiliation(s)
- Yun Tai Kim
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
- Korea Institute of Science and Technology for Eastern Medicine (KISTEM), Neumed Co. Ltd., Seoul 130-701, Korea
| | - Youn-Ju Yi
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Mi-Yeon Kim
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
- Korea Institute of Science and Technology for Eastern Medicine (KISTEM), Neumed Co. Ltd., Seoul 130-701, Korea
| | - Youngmin Bu
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Zhen Hua Jin
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Hoyoung Choi
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
| | - Sylvain Doré
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Hocheol Kim
- Department of Herbal Pharmacology, College of Oriental Medicine, Kyung Hee University, Seoul 130-701, Korea
- Korea Institute of Science and Technology for Eastern Medicine (KISTEM), Neumed Co. Ltd., Seoul 130-701, Korea
| |
Collapse
|
49
|
Pranevicius O, Pranevicius M, Pranevicius H, Liebeskind DS. Transition to collateral flow after arterial occlusion predisposes to cerebral venous steal. Stroke 2012; 43:575-9. [PMID: 22246692 DOI: 10.1161/strokeaha.111.635037] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Stroke-related tissue pressure increase in the core and penumbra determines regional cerebral perfusion pressure (rCPP) defined as a difference between local inflow pressure and venous or tissue pressure, whichever is higher. We previously showed that venous pressure reduction below the pressure in the core causes blood flow diversion-cerebral venous steal. Now we investigated how transition to collateral circulation after complete arterial occlusion affects rCPP distribution. METHODS We modified parallel Starling resistor model to simulate transition to collateral inflow after complete main stem occlusion. We decreased venous pressure from the arterial pressure to zero and investigated how arterial and venous pressure elevation augments rCPP. RESULTS When core pressure exceeded venous, rCPP=inflow pressure in the core. Venous pressure decrease from arterial pressure to pressure in the core caused smaller inflow pressure to drop augmenting rCPP. Further drop of venous pressure decreased rCPP in the core but augmented rCPP in penumbra. After transition to collateral circulation, lowering venous pressure below pressure in the penumbra further decreased rCPP and collaterals themselves became a pathway for steal. Venous pressure level at which rCPP in the core becomes zero we termed the "point of no reflow." Transition from direct to collateral circulation resulted in decreased inflow pressure, decreased rCPP, and a shift of point of no reflow to higher venous loading values. Arterial pressure augmentation increased rCPP, but only after venous pressure exceeded point of no reflow. CONCLUSIONS In the presence of tissue pressure gradients, transition to collateral flow predisposes to venous steal (collateral failure), which may be reversed by venous pressure augmentation.
Collapse
|
50
|
Lee D, Park J, Yoon J, Kim MY, Choi HY, Kim H. Neuroprotective effects of Eleutherococcus senticosus bark on transient global cerebral ischemia in rats. JOURNAL OF ETHNOPHARMACOLOGY 2012; 139:6-11. [PMID: 21645606 DOI: 10.1016/j.jep.2011.05.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 05/11/2011] [Accepted: 05/19/2011] [Indexed: 05/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eleutherococcus senticosus Maxim., classified into the family of Araliaceae, is used in a variety of diseases in traditional Korean medicine including ischemic heart disease. AIM OF THE STUDY To determine the neuroprotective effects of Eleutherococcus senticosus on global cerebral ischemia. MATERIALS AND METHODS A four-vessel occlusion (4-VO) rat model was used to evaluate the potential protective effects against transient global cerebral ischemia ethanol extracts of Eleutherococcus senticosus was orally administered at doses of 3, 30, and 300 mg/kg twice at times of 0 and 90 min after reperfusion. The effects on memory deficit were investigated by using a Y-maze neurobehavioral test after brain ischemia, and the effects on hippocampal neuronal damage were measured 7 days after ischemia. The expressions of glial fibrillary acid protein (GFAP), CD11b antibody (OX-42), and cyclooxygenase-2 (COX-2) were investigated by immunohistochemistry. RESULTS Oral administration of Eleutherococcus seticosus at 30, 100 and 300 mg/kg significantly reduced hippocampal CA1 neuronal death by 3.5%, 25.9% and 53.1%, respectively, compared with a vehicle-treated group. Oral administration of Eleutherococcus senticosus at 300 mg/kg inhibited 81.9% of the decrease in spontaneous alternation induced by 4-VOin the Y-maze test, and also attenuated ischemia-induced activation of COX-2, GFAP and OX-42 in the hippocampal CA1 region. CONCLUSION Eleutherococcus senticosus protects delayed neuronal death in the CA1 region of the hippocampus against global cerebral ischemia in rats with the recovery of spatial memory, which can be considered as the normal functioning of the hippocampus. Regarding the immunohistochemical study, the effect of Eleutherococcus senticosus may be attributable to its anti-inflammatory properties through the inhibition of COX-2 expression, microglia and astrocyte expression.
Collapse
Affiliation(s)
- Donghun Lee
- Department of Herbal Pharmacology, Kyung Hee University, College of Oriental Medicine, Seoul 130-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|