1
|
Su X, Ying J, Xiao D, Qiu X, Li S, Zhao F, Tang J. Activin A rescues preterm brain injury through a novel Noggin/BMP4/Id2 signaling pathway. Int J Mol Med 2022; 51:12. [PMID: 36524372 PMCID: PMC9848437 DOI: 10.3892/ijmm.2022.5215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Activin A (Act A) has been reported to promote oligodendrocyte progenitor cell (OPC) differentiation in vitro and improve neurological outcomes in adult mice. However, the roles and mechanisms of action of Act A in preterm brain injury are unknown. In the present study, P5 rats were subjected to hypoxia‑ischemia to establish a neonatal white matter injury (WMI) model and Act A was injected via the lateral ventricle. Pathological characteristics, OPC differentiation, myelination, and neurological performance were analyzed. Further, the involvement of the Noggin/BMP4/Id2 signaling pathway in the roles of Act A in WMI was explored. Act A attenuated pathological damage, promoted OPC differentiation, enhanced myelin sheath and myelinated axon formation, and improved neurological performance of WMI rats. Moreover, Act A enhanced noggin expression, which, in turn, inhibited the expression of bone morphogenetic protein 4 (BMP4) and inhibitor of DNA binding 2 (Id2). Furthermore, upregulation of Id2 completely abolished the rescue effects of Act A in WMI rats. In conclusion, the present findings suggested that Act A rescues preterm brain injury via targeting a novel Noggin/BMP4/Id2 signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Tang
- Correspondence to: Professor Jun Tang, Department of Pediatrics/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Section 3, 20 South Renmin Road, Chengdu, Sichuan 610041, P.R. China, E-mail:
| |
Collapse
|
2
|
McFarland R, Henzi SP, Fuller A, Hetem RS, Young C, Barrett L. The thermal consequences of primate birth hour and its evolutionary implications. Biol Lett 2022; 18:20210574. [PMID: 35078330 PMCID: PMC8790368 DOI: 10.1098/rsbl.2021.0574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Most primates, including humans, give birth during the inactive phase of the daily cycle. Practical constraints therefore limit our knowledge of the precise timing of nocturnal birth in wild diurnal primates and so limit our understanding of selective pressures and consequences. We measured maternal core body temperature (Tb) across 24 births in a population of wild vervet monkeys using biologgers. We identified distinct perturbations in Tb during the birth period, including declining Tb during labour and the rapid recovery of Tb post-parturition. Vervet monkeys typically gave birth during their inactive phase in synchrony with the nadir of the maternal nychthemeral Tb rhythm but also showed remarkable inter-individual variability in their absolute Tb during birth. Our findings support the view that selection may have favoured a nocturnal timing of primate birth to coincide with lower night-time Tb and environmental temperatures, which improve thermal efficiency during birth.
Collapse
Affiliation(s)
- Richard McFarland
- NTU Psychology, Nottingham Trent University, Nottingham NG1 4FQ, UK,Brain Function Research Group, School of Physiology, Faculty of Health Science, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - S. Peter Henzi
- Department of Psychology, University of Lethbridge, Lethbridge, Canada T1 K 3M4,Applied Behavioural Ecology and Ecosystems Research Unit, University of South Africa, FL 1710, South Africa
| | - Andrea Fuller
- Brain Function Research Group, School of Physiology, Faculty of Health Science, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Robyn S. Hetem
- Brain Function Research Group, School of Physiology, Faculty of Health Science, University of the Witwatersrand, Johannesburg 2193, South Africa,School of Animal, Plant and Environmental Sciences, Faculty of Science, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Christopher Young
- NTU Psychology, Nottingham Trent University, Nottingham NG1 4FQ, UK,Applied Behavioural Ecology and Ecosystems Research Unit, University of South Africa, FL 1710, South Africa
| | - Louise Barrett
- Brain Function Research Group, School of Physiology, Faculty of Health Science, University of the Witwatersrand, Johannesburg 2193, South Africa,Department of Psychology, University of Lethbridge, Lethbridge, Canada T1 K 3M4
| |
Collapse
|
3
|
ARAL AL, ERGÜN MA, ENGİN AB, BÖRCEK AÖ, BOLAY H. Iron homeostasis is altered in response to hypoxia and hypothermic preconditioning in brain glial cells. Turk J Med Sci 2020; 50:2005-2016. [PMID: 32682355 PMCID: PMC7775693 DOI: 10.3906/sag-2003-41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022] Open
Abstract
Background/aim Altered iron metabolism is one of the pathophysiological mechanisms occurring during hypoxic injuries in the central nervous system. Proper homeostasis of cellular iron is regulated by iron import, storage, and export proteins that prevent excess iron overload or iron starvation in cells. Therapeutic hypothermia is an approved treatment for hypoxic ischemia in newborns, but the underlying molecular mechanism is still unknown. We studied the effects of hypoxia, preceded with preconditioning, on the iron homeostasis of glial cells, known as a major actor in the inflammatory process during perinatal brain injury. Materials and methods Primary microglia and astrocytes in culture were exposed to 12 h of hypoxia with or without mild hypothermic preconditioning. The mRNA expression was assessed using qPCR. Iron accumulation was visualized via modified Perl’s histochemistry. Cytokine levels in cell cultures were measured using ELISA. Results Hypothermic preconditioning enhanced microglial viability, which previously was decreased in both cell types due to hypoxia. Hypoxia increased iron accumulation in the mixed glial cells and in ferritin expression in both microglia and astrocytes. Hypotermic preconditioning decreased the elevated ferritin-light chain expression significantly in microglia. Iron importer proteins, DMT1 and TfR1, both increased their mRNA expression after hypoxia, and hypothermic preconditioning continued to support the elevation of DMT1 in both glial cell types. Ferroportin expression increased as a survival factor of the glial cell following hypoxia. Hypothermic preconditioning supported this increase in both cell types and was especially significant in astrocytes. IL-10 levels were prominently increased in cell culture after hypothermic preconditioning. Conclusion The data suggest that hypothermic preconditioning affects cellular iron homeostasis by regulating the storage and transfer proteins of iron. Regulation of the cellular iron traffic may prevent glial cells from experiencing the detrimental effects of hypoxia-related inflammation.
Collapse
Affiliation(s)
- Arzu L. ARAL
- Department of Immunology, Faculty of Medicine, İzmir Demokrasi University, İzmirTurkey
| | - Mehmet Ali ERGÜN
- Department of Genetics, Faculty of Medicine, Gazi University, AnkaraTurkey
| | - Ayşe Başak ENGİN
- Department of Toxicology, Faculty of Pharmacy, Gazi University, AnkaraTurkey
| | - Alp Özgün BÖRCEK
- Department of Neurosurgery, Faculty of Medicine, Gazi University, AnkaraTurkey
| | - Hayrunnisa BOLAY
- Department of Neurology, Faculty of Medicine, Gazi University, AnkaraTurkey
| |
Collapse
|
4
|
Hao Y, Xin M, Feng L, Wang X, Wang X, Ma D, Feng J. Review Cerebral Ischemic Tolerance and Preconditioning: Methods, Mechanisms, Clinical Applications, and Challenges. Front Neurol 2020; 11:812. [PMID: 33071923 PMCID: PMC7530891 DOI: 10.3389/fneur.2020.00812] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of morbidity and mortality worldwide, and it is increasing in prevalence. The limited therapeutic window and potential severe side effects prevent the widespread clinical application of the venous injection of thrombolytic tissue plasminogen activator and thrombectomy, which are regarded as the only approved treatments for acute ischemic stroke. Triggered by various types of mild stressors or stimuli, ischemic preconditioning (IPreC) induces adaptive endogenous tolerance to ischemia/reperfusion (I/R) injury by activating a multitude cascade of biomolecules, for example, proteins, enzymes, receptors, transcription factors, and others, which eventually lead to transcriptional regulation and epigenetic and genomic reprogramming. During the past 30 years, IPreC has been widely studied to confirm its neuroprotection against subsequent I/R injury, mainly including local ischemic preconditioning (LIPreC), remote ischemic preconditioning (RIPreC), and cross preconditioning. Although LIPreC has a strong neuroprotective effect, the clinical application of IPreC for subsequent cerebral ischemia is difficult. There are two main reasons for the above result: Cerebral ischemia is unpredictable, and LIPreC is also capable of inducing unexpected injury with only minor differences to durations or intensity. RIPreC and pharmacological preconditioning, an easy-to-use and non-invasive therapy, can be performed in a variety of clinical settings and appear to be more suitable for the clinical management of ischemic stroke. Hoping to advance our understanding of IPreC, this review mainly focuses on recent advances in IPreC in stroke management, its challenges, and the potential study directions.
Collapse
Affiliation(s)
| | | | | | | | | | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Jiang L, Wan W, Xun Y, Xiong L, Wu B, Xiang Y, Li Z, Zhu L, Ji Y, Yang P, Hu K. Effect of hypothermic perfusion on phacoemulsification in cataract patients complicated with uveitis: a randomised trial. BMC Ophthalmol 2020; 20:232. [PMID: 32546211 PMCID: PMC7298810 DOI: 10.1186/s12886-020-01507-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 06/05/2020] [Indexed: 11/18/2022] Open
Abstract
Background To evaluate the effectiveness and safety of hypothermic perfusion in the phacoemulsification of cataract caused by uveitis. Methods This was a prospective, single-masked, randomised, controlled clinical trial. One hundred and six patients with uveitis-associated cataract underwent phacoemulsification with perfusion fluid temperature at 4 °C (treatment group) or 24 °C (control group). Anterior chamber inflammation grade, corneal endothelial cell count, corneal thickness, macular fovea thickness, and intraocular pressure (IOP) were observed on the 1st day and 7th day after operation. Results The aqueous flare score was 0.83 ± 0.76 in the 4 °C group, which was lower than that in the 24 °C group (1.51 ± 1.02, p = 0.006) on the first day after operation. The aqueous cells score was lower in the 4 °C group (0.17 ± 0.38) than that in the 24 °C group (0.62 ± 0.94, p = 0.025). The mean corneal thickness of incision in the 4 °C group (907.66 ± 85.37 μm) was thinner than that in the 24 °C group (963.75 ± 103.81 μm, p = 0.005). Corneal endothelial cells density, macular fovea thickness, or percentage of transiently increased IOP showed no difference between the two groups (p > 0.05). There was no significant difference in all the main outcome parameters between the two groups on the 7th day after operation (p > 0.05). Conclusions Hypothermic perfusion in the phacoemulsification of uveitis-associated cataract is safe, and it can effectively inhibit anterior chamber inflammation and reduce the incisional corneal edema in the early postoperative stage. Trial registration The study was registered with the Chinese Clinical Trial Registry. (http://www.chictr.org.cn/, Registration Number: ChiCTR1800016145).
Collapse
Affiliation(s)
- Lu Jiang
- Chongqing Medical University, Chongqing, China
| | - Wenjuan Wan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Yan Xun
- Chongqing Medical University, Chongqing, China
| | - Liang Xiong
- Chongqing Medical University, Chongqing, China
| | - Binge Wu
- The Second affiliated hospital of Baotou medical college, Baotou, Inner Mongolia Autonomous Region, China
| | | | - Zhouyu Li
- Chongqing Medical University, Chongqing, China
| | - Lu Zhu
- Chongqing Medical University, Chongqing, China
| | - Yan Ji
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Ke Hu
- Chongqing Medical University, Chongqing, China. .,The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China.
| |
Collapse
|
6
|
Wan W, Jiang L, Ji Y, Xun Y, Xiong L, Xiang Y, Li R, Li Z, Wang X, Stewart JM, Hu K. Effect of hypothermic perfusion on phacoemulsification in eyes with hard nuclear cataract: randomized trial. J Cataract Refract Surg 2019; 45:1717-1724. [PMID: 31856981 DOI: 10.1016/j.jcrs.2019.07.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023]
Abstract
PURPOSE To evaluate the effectiveness and safety of hypothermic perfusion in the phacoemulsification of hard nuclear cataract. SETTING Tertiary opthalmology center, China. DESIGN Laboratory study and prospective randomized clinical trial. METHODS Rabbits and patients with hard nuclear cataract underwent phacoemulsification with perfusion temperatures at 4°C or 24°C. Anterior segment optical coherence tomography (AS-OCT), corneal endothelial cell count (ECC), and cornea sections were observed before the rabbits' operation and 1 day and 7 days postoperatively. AS-OCT, corneal ECC, and anterior chamber (AC) inflammation were observed before the patients' operation and 1 day, 7 days, and 30 days postoperatively. RESULTS The study comprised 40 rabbits and 80 patients. In the animal models, the mean central corneal thickness (CCT) in the 4°C group (370.4 μm ± 45.5 [SD]) was thinner than in the 24°C group (496.7 ± 121.5 μm) 1 day postoperatively (P < .001). The mean AC inflammation reaction grade in the 4°C group (1.1 ± 0.9) was lower than in the 24°C group (2.2 ± 0.8) (P = .0333). In clinical trials, the mean CCT and incisional corneal thicknesses in the 4°C group (600.7 ± 51.8 μm and 859.2 ± 177.8 μm, respectively) were thinner than in the 24°C group (655.3 ± 85.0 μm and 955.9 ± 196.7 μm, respectively) (P < .001). The endothelial cell density (P = .036) and hexagonality (P = .001) were higher in the 4°C group. The mean AC inflammation reaction grade in the 4°C group (0.6 ± 0.6) was lower than in the 24°C group (1.3 ± 1.0) 1 day postoperatively (P = .004). CONCLUSIONS Hypothermic perfusion in phacoemulsification of hard nuclear cataract is safe and it can effectively protect corneal endothelium, decrease corneal edema, and reduce AC inflammation in the early postoperative stage.
Collapse
Affiliation(s)
- Wenjuan Wan
- First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Lu Jiang
- Chongqing Medical University, Chongqing, China
| | - Yan Ji
- First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Yan Xun
- Chongqing Medical University, Chongqing, China
| | - Liang Xiong
- Chongqing Medical University, Chongqing, China
| | | | - Ruonan Li
- Chongqing Medical University, Chongqing, China
| | - Zhouyu Li
- Chongqing Medical University, Chongqing, China
| | - Xiaoqin Wang
- People's Hospital of Tongliang District, Chongqing, China
| | - Jay M Stewart
- Department of Ophthalmology, University of California, San Francisco, USA
| | - Ke Hu
- First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China.
| |
Collapse
|
7
|
Kreisman NR, Wooliscroft LB, Campbell CF, Dotiwala AK, Cox ML, Denson AC, Betancourt AM, Tomchuck SL. Preconditioning hippocampal slices with hypothermia promotes rapid tolerance to hypoxic depolarization and swelling: Mediation by erythropoietin. Brain Res 2019; 1726:146517. [PMID: 31634451 DOI: 10.1016/j.brainres.2019.146517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/06/2019] [Accepted: 10/17/2019] [Indexed: 12/26/2022]
Abstract
We suggested previously that hippocampal slices were protected from hypoxic depolarization and swelling by preincubating them at room temperature (Kreisman et al., 2000). We postulated that hypothermic preconditioning induced tolerance in our slices, which protected against hypoxic depolarization and swelling. Control hippocampal slices were incubated at 34-35 °C for two hours and the response to 10 min of severe hypoxia was compared to slices which were preconditioned for two hours at room temperature (22-23 °C) prior to warming to 34-35 °C. Recordings of the extracellular DC potential provided an index of tissue depolarization and changes in tissue light transmittance provided an index of swelling. Hypothermic preconditioning significantly reduced hypoxia-induced swelling, particularly in CA3 and the dentate inner blade. Since erythropoietin (EPO) had been shown to mediate hypoxic preconditioning, we tested whether EPO also mediated hypothermic preconditioning in our slices. Recombinant rat EPO (1-10 micromolar) mitigated hypoxia-induced swelling and depolarization in dentate inner blade of unconditioned slices in a dose-dependent manner. We also blocked the protective effects of hypothermic preconditioning on hypoxic depolarization and swelling in the inner blade of the dentate gyrus by administering soluble EPO receptor in the bath and treating slices with wortmannin to block phosphorylation of PI3 kinase, a critical step in the activation of the downstream neuroprotectant, Akt. These results suggest that EPO mediates tolerance to hypoxic depolarization and swelling induced by hypothermic preconditioning. They also emphasize that various preincubation protocols used in experiments with hippocampal slices may differentially affect basal electrophysiological and metabolic properties of those slices.
Collapse
Affiliation(s)
- Norman R Kreisman
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Tulane Brain Institute, New Orleans, LA 70118, United States.
| | | | - Carolyn F Campbell
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Ary K Dotiwala
- Tulane Brain Institute, New Orleans, LA 70118, United States
| | - Michael L Cox
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aaron C Denson
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aline M Betancourt
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Suzanne L Tomchuck
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
8
|
Su X, Huang L, Xiao D, Qu Y, Mu D. Research Progress on the Role and Mechanism of Action of Activin A in Brain Injury. Front Neurosci 2018; 12:697. [PMID: 30356877 PMCID: PMC6190887 DOI: 10.3389/fnins.2018.00697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/18/2018] [Indexed: 12/16/2022] Open
Abstract
Activin A belongs to the transforming growth factor superfamily and has a variety of biological functions. Studies have revealed that activin A can regulate the body's immune and inflammatory responses and participate in the regulation of cell death. In addition, activin A also has neurotrophic function and plays an important role in the repair of brain damage. This article summarizes recent advances in understanding the role and mechanism of action of activin A in brain injury and provides new hints into the application of activin A in the treatment of brain injury.
Collapse
Affiliation(s)
- Xiaojuan Su
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Lingyi Huang
- Department of Stomatology, West China College of Stomatology, Sichuan University, Chengdu, China
| | - Dongqiong Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
9
|
Vinciguerra A, Cuomo O, Cepparulo P, Anzilotti S, Brancaccio P, Sirabella R, Guida N, Annunziato L, Pignataro G. Models and methods for conditioning the ischemic brain. J Neurosci Methods 2018; 310:63-74. [PMID: 30287283 DOI: 10.1016/j.jneumeth.2018.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/13/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND In the last decades the need to find new neuroprotective targets has addressed the researchers to investigate the endogenous molecular mechanisms that brain activates when exposed to a conditioning stimulus. Indeed, conditioning is an adaptive biological process activated by those interventions able to confer resistance to a deleterious brain event through the exposure to a sub-threshold insult. Specifically, preconditioning and postconditioning are realized when the conditioning stimulus is applied before or after, respectively, the harmul ischemia. AIMS AND RESULTS The present review will describe the most common methods to induce brain conditioning, with particular regards to surgical, physical exercise, temperature-induced and pharmacological approaches. It has been well recognized that when the subliminal stimulus is delivered after the ischemic insult, the achieved neuroprotection is comparable to that observed in models of ischemic preconditioning. In addition, subjecting the brain to both preconditioning as well as postconditioning did not cause greater protection than each treatment alone. CONCLUSIONS The last decades have provided fascinating insights into the mechanisms and potential application of strategies to induce brain conditioning. Since the identification of intrinsic cell-survival pathways should provide more direct opportunities for translational neuroprotection trials, an accurate examination of the different models of preconditioning and postconditioning is mandatory before starting any new project.
Collapse
Affiliation(s)
- Antonio Vinciguerra
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | - Pasquale Cepparulo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | | | - Paola Brancaccio
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy
| | | | | | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, School of Medicine, "Federico II" University of Naples, Via Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
10
|
Yunoki M, Kanda T, Suzuki K, Uneda A, Hirashita K, Yoshino K. Ischemic Tolerance of the Brain and Spinal Cord: A Review. Neurol Med Chir (Tokyo) 2017; 57:590-600. [PMID: 28954945 PMCID: PMC5709712 DOI: 10.2176/nmc.ra.2017-0062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ischemic tolerance is an endogenous neuroprotective phenomenon induced by sublethal ischemia. Ischemic preconditioning (IPC), the first discovered form of ischemic tolerance, is widely seen in many species and in various organs including the brain and the spinal cord. Ischemic tolerance of the spinal cord is less familiar among neurosurgeons, although it has been reported from the viewpoint of preventing ischemic spinal cord injury during aortic surgery. It is important for neurosurgeons to have opportunities to see patients with spinal cord ischemia, and to understand ischemic tolerance of the spinal cord as well as the brain. IPC has a strong neuroprotective effect in animal models of ischemia; however, clinical application of IPC for ischemic brain and spinal diseases is difficult because they cannot be predicted. In addition, one drawback of preconditioning stimuli is that they are also capable of producing injury with only minor changes to their intensity or duration. Numerous methods to induce ischemic tolerance have been discovered that vary in their timing and the site at which short-term ischemia occurs. These methods include ischemic postconditioning (IPoC), remote ischemic preconditioning (RIPC), remote ischemic perconditioning (RIPerC) and remote ischemic postconditioning (RIPoC), which has had a great impact on clinical approaches to treatment of ischemic brain and spinal cord injury. Especially RIPerC and RIPoC to induce spinal cord tolerance are considered clinically useful, however the evidence supporting these methods is currently insufficient; further experimental or clinical research in this area is thus necessary.
Collapse
Affiliation(s)
| | | | - Kenta Suzuki
- Department of Neurosurgery, Kagawa Rosai Hospital
| | | | | | | |
Collapse
|
11
|
Postconditioning-induced neuroprotection, mechanisms and applications in cerebral ischemia. Neurochem Int 2017; 107:43-56. [DOI: 10.1016/j.neuint.2017.01.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/04/2017] [Accepted: 01/08/2017] [Indexed: 02/07/2023]
|
12
|
Dhillon SK, Gunn AJ, Jung Y, Mathai S, Bennet L, Fraser M. Lipopolysaccharide-Induced Preconditioning Attenuates Apoptosis and Differentially Regulates TLR4 and TLR7 Gene Expression after Ischemia in the Preterm Ovine Fetal Brain. Dev Neurosci 2015; 37:497-514. [PMID: 26184807 DOI: 10.1159/000433422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/13/2015] [Indexed: 11/19/2022] Open
Abstract
Acute exposure to subclinical infection modulates subsequent hypoxia-ischemia (HI) injury in a time-dependent manner, likely by cross-talk through Toll-like receptors (TLRs), but the specific pathways are unclear in the preterm-equivalent brain. In the present study, we tested the hypothesis that repeated low-dose exposure to lipopolysaccharide (LPS) before acute ischemia would be associated with induction of specific TLRs that are potentially neuroprotective. Fetal sheep at 0.65 gestation (term is ∼145 days) received intravenous boluses of low-dose LPS for 5 days (day 1, 50 ng/kg; days 2-5, 100 ng/kg) or the same volume of saline. Either 4 or 24 h after the last bolus of LPS, complete carotid occlusion was induced for 22 min. Five days after LPS, brains were collected. Pretreatment with LPS for 5 days decreased cellular apoptosis, microglial activation and reactive astrogliosis in response to HI injury induced 24 but not 4 h after the last dose of LPS. This was associated with upregulation of TLR4, TLR7 and IFN-β mRNA, and increased fetal plasma IFN-β concentrations. The association of reduced white matter apoptosis and astrogliosis after repeated low-dose LPS finishing 24 h but not 4 h before cerebral ischemia, with central and peripheral induction of IFN-β, suggests the possibility that IFN-β may be an important mediator of endogenous neuroprotection in the developing brain.
Collapse
|
13
|
Thushara Vijayakumar N, Sangwan A, Sharma B, Majid A, Rajanikant GK. Cerebral Ischemic Preconditioning: the Road So Far…. Mol Neurobiol 2015; 53:2579-93. [PMID: 26081149 DOI: 10.1007/s12035-015-9278-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 06/02/2015] [Indexed: 12/25/2022]
Abstract
Cerebral preconditioning constitutes the brain's adaptation to lethal ischemia when first exposed to mild doses of a subtoxic stressor. The phenomenon of preconditioning has been largely studied in the heart, and data from in vivo and in vitro models from past 2-3 decades have provided sufficient evidence that similar machinery exists in the brain as well. Since preconditioning results in a transient protective phenotype labeled as ischemic tolerance, it can open many doors in the medical warfare against stroke, a debilitating cerebrovascular disorder that kills or cripples thousands of people worldwide every year. Preconditioning can be induced by a variety of stimuli from hypoxia to pharmacological anesthetics, and each, in turn, induces tolerance by activating a multitude of proteins, enzymes, receptors, transcription factors, and other biomolecules eventually leading to genomic reprogramming. The intracellular signaling pathways and molecular cascades behind preconditioning are extensively being investigated, and several first-rate papers have come out in the last few years centered on the topic of cerebral ischemic tolerance. However, translating the experimental knowledge into the clinical scaffold still evades practicality and faces several challenges. Of the various preconditioning strategies, remote ischemic preconditioning and pharmacological preconditioning appears to be more clinically relevant for the management of ischemic stroke. In this review, we discuss current developments in the field of cerebral preconditioning and then examine the potential of various preconditioning agents to confer neuroprotection in the brain.
Collapse
Affiliation(s)
- N Thushara Vijayakumar
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Amit Sangwan
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Bhargy Sharma
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Arshad Majid
- Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - G K Rajanikant
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
14
|
Hypothermic Preconditioning of Human Cortical Neurons Requires Proteostatic Priming. EBioMedicine 2015; 2:528-35. [PMID: 26287272 PMCID: PMC4534756 DOI: 10.1016/j.ebiom.2015.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/06/2015] [Accepted: 04/08/2015] [Indexed: 01/23/2023] Open
Abstract
Hypothermia is potently neuroprotective but poor mechanistic understanding has restricted its clinical use. Rodent studies indicate that hypothermia can elicit preconditioning, wherein a subtoxic cellular stress confers resistance to an otherwise lethal injury. The molecular basis of this preconditioning remains obscure. Here we explore molecular effects of cooling using functional cortical neurons differentiated from human pluripotent stem cells (hCNs). Mild-to-moderate hypothermia (28–32 °C) induces cold-shock protein expression and mild endoplasmic reticulum (ER) stress in hCNs, with full activation of the unfolded protein response (UPR). Chemical block of a principal UPR pathway mitigates the protective effect of cooling against oxidative stress, whilst pre-cooling neurons abrogates the toxic injury produced by the ER stressor tunicamycin. Cold-stress thus preconditions neurons by upregulating adaptive chaperone-driven pathways of the UPR in a manner that precipitates ER-hormesis. Our findings establish a novel arm of neurocryobiology that could reveal multiple therapeutic targets for acute and chronic neuronal injury. Clinically-relevant cooling induces archetypal cold-shock and mild endoplasmic reticulum (ER) stress in human neurons. Hypothermic neuronal ER-stress elicits an adaptive unfolded protein response (UPR) with ER-hormesis. Hypothermic preconditioning of the ER provides cross-tolerance to oxidative neuronal injury and requires an intact UPR.
Collapse
|
15
|
Lee YJ, Mou Y, Klimanis D, Bernstock JD, Hallenbeck JM. Global SUMOylation is a molecular mechanism underlying hypothermia-induced ischemic tolerance. Front Cell Neurosci 2014; 8:416. [PMID: 25538566 PMCID: PMC4255597 DOI: 10.3389/fncel.2014.00416] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/16/2014] [Indexed: 12/20/2022] Open
Abstract
The molecular mechanisms underlying hypothermic neuroprotection have yet to be fully elucidated. Herein we demonstrate that global SUMOylation, a form of post-translational modification with the Small Ubiquitin-like MOdifer, participates in the multimodal molecular induction of hypothermia-induced ischemic tolerance. Mild (32°C) to moderate (28°C) hypothermic treatment(s) during OGD (oxygen-glucose-deprivation) or ROG (restoration of oxygen/glucose) increased global SUMO-conjugation levels and protected cells (both SHSY5Y and E18 rat cortical neurons) from OGD and ROG-induced cell death. Hypothermic exposure either before or after permanent middle cerebral artery occlusion (pMCAO) surgery in wild type mice increased global SUMO-conjugation levels in the brain and in so doing protected these animals from pMCAO-induced ischemic damage. Of note, hypothermic exposure did not provide an additional increase in protection from pMCAO-induced ischemic brain damage in Ubc9 transgenic (Ubc9 Tg) mice, which overexpress the sole E2 SUMO conjugating enzyme and thereby display elevated basal levels of global SUMOylation under normothermic conditions. Such evidence suggests that increases in global SUMOylation are critical and may account for a substantial part of the observed increase in cellular tolerance to brain ischemia caused via hypothermia.
Collapse
Affiliation(s)
- Yang-Ja Lee
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health Bethesda, MD, USA
| | - Yongshan Mou
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health Bethesda, MD, USA
| | - Dace Klimanis
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health Bethesda, MD, USA
| | - Joshua D Bernstock
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health Bethesda, MD, USA
| | - John M Hallenbeck
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
16
|
Leandra C, Tasca CI, Boeck CR. The Role of NMDA Receptors in the Development of Brain Resistance through Pre- and Postconditioning. Aging Dis 2014; 5:430-41. [PMID: 25489494 DOI: 10.14336/ad.2014.0500430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/30/2014] [Accepted: 02/09/2014] [Indexed: 11/01/2022] Open
Abstract
Brain tolerance or resistance can be achieved by interventions before and after injury through potential toxic agents used in low stimulus or dose. For brain diseases, the neuroprotection paradigm desires an attenuation of the resulting motor, cognitive, emotional, or memory deficits following the insult. Preconditioning is a well-established experimental and clinical translational strategy with great beneficial effects, but limited applications. NMDA receptors have been reported as protagonists in the adjacent cellular mechanisms contributing to the development of brain tolerance. Postconditioning has recently emerged as a new neuroprotective strategy, which has shown interesting results when applied immediately, i.e. several hours to days, after a stroke event. Investigations using chemical postconditioning are still incipient, but nevertheless represent an interesting and promising clinical strategy. In the present review pre- and postconditioning are discussed as neuroprotective paradigms and the focus of our attention lies on the participation of NMDA receptors proteins in the processes related to neuroprotection.
Collapse
Affiliation(s)
| | - Carla Inês Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Carina Rodrigues Boeck
- Laboratório de Biologia Celular e Molecular, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-graduação Ciências da Saúde, Universidade do Extremo Sul Catarinense-UNESC, Criciúma, 88806-000, SC, Brazil
| |
Collapse
|
17
|
Vitreous humor thermodynamics during phacoemulsification. Int Ophthalmol 2014; 35:557-64. [DOI: 10.1007/s10792-014-9983-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/26/2014] [Indexed: 11/26/2022]
|
18
|
Ma XD, Song JN, Zhang M, An JY, Zhao YL, Zhang BF. Advances in research of the neuroprotective mechanisms of cerebral ischemic postconditioning. Int J Neurosci 2014; 125:161-9. [PMID: 24754439 DOI: 10.3109/00207454.2014.917413] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Ischemic postconditioning refers to controlling reperfusion blood flow during reperfusion after ischemia, which can induce an endogenous neuroprotective effect and reduce ischemia-reperfusion injury. Activation of endogenous neuroprotective mechanisms plays a key role in protecting against brain ischemia-reperfusion injury. The mechanisms of cerebral ischemic postconditioning are not completely clear, and the following aspects may be involved: downregulation of oxidative stress, attenuating mitochondrial dysfunction, attenuating endoplasmic reticulum stress, accelerating the elimination of glutamate, increasing rCBF, inhibiting apoptosis, inhibiting autophagy, and regulating signal transduction.
Collapse
Affiliation(s)
- Xu-Dong Ma
- Department of Neurosurgery, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | |
Collapse
|
19
|
Garcia-Bonilla L, Benakis C, Moore J, Iadecola C, Anrather J. Immune mechanisms in cerebral ischemic tolerance. Front Neurosci 2014; 8:44. [PMID: 24624056 PMCID: PMC3940969 DOI: 10.3389/fnins.2014.00044] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 02/17/2014] [Indexed: 12/21/2022] Open
Abstract
Stressor-induced tolerance is a central mechanism in the response of bacteria, plants, and animals to potentially harmful environmental challenges. This response is characterized by immediate changes in cellular metabolism and by the delayed transcriptional activation or inhibition of genetic programs that are not generally stressor specific (cross-tolerance). These programs are aimed at countering the deleterious effects of the stressor. While induction of this response (preconditioning) can be established at the cellular level, activation of systemic networks is essential for the protection to occur throughout the organs of the body. This is best signified by the phenomenon of remote ischemic preconditioning, whereby application of ischemic stress to one tissue or organ induces ischemic tolerance (IT) in remote organs through humoral, cellular and neural signaling. The immune system is an essential component in cerebral IT acting simultaneously both as mediator and target. This dichotomy is based on the fact that activation of inflammatory pathways is necessary to establish IT and that IT can be, in part, attributed to a subdued immune activation after index ischemia. Here we describe the components of the immune system required for induction of IT and review the mechanisms by which a reprogrammed immune response contributes to the neuroprotection observed after preconditioning. Learning how local and systemic immune factors participate in endogenous neuroprotection could lead to the development of new stroke therapies.
Collapse
Affiliation(s)
- Lidia Garcia-Bonilla
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Corinne Benakis
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Jamie Moore
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Costantino Iadecola
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Josef Anrather
- Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| |
Collapse
|
20
|
Stetler RA, Leak RK, Gan Y, Li P, Zhang F, Hu X, Jing Z, Chen J, Zigmond MJ, Gao Y. Preconditioning provides neuroprotection in models of CNS disease: paradigms and clinical significance. Prog Neurobiol 2014; 114:58-83. [PMID: 24389580 PMCID: PMC3937258 DOI: 10.1016/j.pneurobio.2013.11.005] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/14/2022]
Abstract
Preconditioning is a phenomenon in which brief episodes of a sublethal insult induce robust protection against subsequent lethal injuries. Preconditioning has been observed in multiple organisms and can occur in the brain as well as other tissues. Extensive animal studies suggest that the brain can be preconditioned to resist acute injuries, such as ischemic stroke, neonatal hypoxia/ischemia, surgical brain injury, trauma, and agents that are used in models of neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. Effective preconditioning stimuli are numerous and diverse, ranging from transient ischemia, hypoxia, hyperbaric oxygen, hypothermia and hyperthermia, to exposure to neurotoxins and pharmacological agents. The phenomenon of "cross-tolerance," in which a sublethal stress protects against a different type of injury, suggests that different preconditioning stimuli may confer protection against a wide range of injuries. Research conducted over the past few decades indicates that brain preconditioning is complex, involving multiple effectors such as metabolic inhibition, activation of extra- and intracellular defense mechanisms, a shift in the neuronal excitatory/inhibitory balance, and reduction in inflammatory sequelae. An improved understanding of brain preconditioning should help us identify innovative therapeutic strategies that prevent or at least reduce neuronal damage in susceptible patients. In this review, we focus on the experimental evidence of preconditioning in the brain and systematically survey the models used to develop paradigms for neuroprotection, and then discuss the clinical potential of brain preconditioning.
Collapse
Affiliation(s)
- R Anne Stetler
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Yu Gan
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Peiying Li
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Feng Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoming Hu
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Zheng Jing
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Michael J Zigmond
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China.
| |
Collapse
|
21
|
Nunes RR, Duval Neto GF, Garcia de Alencar JC, Franco SB, de Andrade NQ, Holanda Dumaresq DM, Cavalcante SL. Anesthetics, cerebral protection and preconditioning. Rev Bras Anestesiol 2014; 63:119-28. [PMID: 23438807 DOI: 10.1016/s0034-7094(13)70204-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 06/16/2012] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Several studies demonstrate that cerebral preconditioning is a protective mechanism against a stressful situation. Preconditioning determinants are described, as well as the neuroprotection provided by anesthetic and non-anesthetics agents. CONTENT Review based on the main articles addressing the pathophysiology of ischemia-reperfusion and neuronal injury and pharmacological and non-pharmacological factors (inflammation, glycemia, and temperature) related to the change in response to ischemia-reperfusion, in addition to neuroprotection induced by anesthetic use. CONCLUSIONS The brain has the ability to protect itself against ischemia when stimulated. The elucidation of this mechanism enables the application of preconditioning inducing substances (some anesthetics), other drugs, and non-pharmacological measures, such as hypothermia, aimed at inducing tolerance to ischemic lesions.
Collapse
|
22
|
Nunes RR, Duval Neto GF, de Alencar JCG, Franco SB, de Andrade NQ, Dumaresq DMH, Cavalcante SL. Anesthetics, cerebral protection and preconditioning. Braz J Anesthesiol 2013; 63:119-28. [PMID: 24565096 DOI: 10.1016/j.bjane.2012.06.003] [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] [Received: 10/14/2011] [Accepted: 06/16/2012] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Several studies demonstrate that cerebral preconditioning is a protective mechanism against a stressful situation. Preconditioning determinants are described, as well as the neuroprotection provided by anesthetic and non-anesthetics agents. CONTENT Review based on the main articles addressing the pathophysiology of ischemia-reperfusion and neuronal injury and pharmacological and non-pharmacological factors (inflammation, glycemia, and temperature) related to the change in response to ischemia-reperfusion, in addition to neuroprotection induced by anesthetic use. CONCLUSIONS The brain has the ability to protect itself against ischemia when stimulated. The elucidation of this mechanism enables the application of preconditioning inducing substances (some anesthetics), other drugs, and non-pharmacological measures, such as hypothermia, aimed at inducing tolerance to ischemic lesions.
Collapse
Affiliation(s)
- Rogean Rodrigues Nunes
- TSA; MSc and PhD in Anesthetics; Postgraduate in Cardiology, Universidade Federal do Ceará (UFC); Jointly Responsible for the Center for Teaching and Training (CET) of Hospital Geral de Fortaleza (HGF); Medicine Professor of Fachristus; Postgraduate in Clinical Engineering, Universidade de Fortaleza (Unifor); Vice-Chair of the Research Ethics Committee, Hospital São Carlos, Fortaleza, Ceará.
| | | | | | | | | | - Danielle Maia Holanda Dumaresq
- TSA; MSc, UFC; Responsible for CET-IJF; Chairman of the Pediatric Anesthesia Committee, Sociedade Brasileira de Anestesiologia (SBA) - 2011; Medicine Professor of Fachristus, Fortaleza, Ceará
| | - Sara Lúcia Cavalcante
- PhD; Professor, Faculdade de Medicina, UFC; Corresponsible for the CET of the HGF from Hospital São Carlos, Fortaleza Ceara, Brazil
| |
Collapse
|
23
|
Salido EM, Dorfman D, Bordone M, Chianelli M, González Fleitas MF, Rosenstein RE. Global and ocular hypothermic preconditioning protect the rat retina from ischemic damage. PLoS One 2013; 8:e61656. [PMID: 23626711 PMCID: PMC3633982 DOI: 10.1371/journal.pone.0061656] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/12/2013] [Indexed: 11/18/2022] Open
Abstract
Retinal ischemia could provoke blindness. At present, there is no effective treatment against retinal ischemic damage. Strong evidence supports that glutamate is implicated in retinal ischemic damage. We investigated whether a brief period of global or ocular hypothermia applied 24 h before ischemia (i.e. hypothermic preconditioning, HPC) protects the retina from ischemia/reperfusion damage, and the involvement of glutamate in the retinal protection induced by HPC. For this purpose, ischemia was induced by increasing intraocular pressure to 120 mm Hg for 40 min. One day before ischemia, animals were submitted to global or ocular hypothermia (33°C and 32°C for 20 min, respectively) and fourteen days after ischemia, animals were subjected to electroretinography and histological analysis. Global or ocular HPC afforded significant functional (electroretinographic) protection in eyes exposed to ischemia/reperfusion injury. A marked alteration of the retinal structure and a decrease in retinal ganglion cell number were observed in ischemic retinas, whereas global or ocular HPC significantly preserved retinal structure and ganglion cell count. Three days after ischemia, a significant decrease in retinal glutamate uptake and glutamine synthetase activity was observed, whereas ocular HPC prevented the effect of ischemia on these parameters. The intravitreal injection of supraphysiological levels of glutamate induced alterations in retinal function and histology which were significantly prevented by ocular HPC. These results support that global or ocular HPC significantly protected retinal function and histology from ischemia/reperfusion injury, probably through a glutamate-dependent mechanism.
Collapse
Affiliation(s)
- Ezequiel M Salido
- Laboratory of Retinal Neurochemistry and Experimental Ophthalmology, Department of Human Biochemistry, School of Medicine, University of Buenos Aires/CEFyBO, CONICET, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
24
|
Mou Y, Wilgenburg BJ, Lee YJ, Hallenbeck JM. A method for hypothermia-induction and maintenance allows precise body and brain temperature control in mice. J Neurosci Methods 2012; 213:1-5. [PMID: 23174093 DOI: 10.1016/j.jneumeth.2012.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 11/07/2012] [Accepted: 11/08/2012] [Indexed: 12/01/2022]
Abstract
The benefits as well as mechanisms of hypothermia in brain injuries are actively studied at the bench and in the clinic. However, methods used in controlling hypothermia vary among laboratories, and usually brain temperatures are not monitored directly in animals due to the need for an invasive procedure. Here we show a method, water immersion technique, which we developed recently to regulate body temperature in mice during hypothermia process. This method significantly reduced the temperature variation around target temperature. Importantly, this method demonstrated a parallel and consistent relationship between rectal temperature and brain temperature (the brain temperature was consistently 0.5C higher than rectal temperature) throughout hypothermia maintenance. This technique may be well adapted to hypothermia studies in mice and other rodents, especially to the assessment and regulation of brain temperature during studies.
Collapse
Affiliation(s)
- Yongshan Mou
- Clinical Investigations Section, Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
25
|
Wang L, Ma Q, Yang W, Mackensen GB, Paschen W. Moderate hypothermia induces marked increase in levels and nuclear accumulation of SUMO2/3-conjugated proteins in neurons. J Neurochem 2012; 123:349-59. [PMID: 22891650 DOI: 10.1111/j.1471-4159.2012.07916.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/01/2012] [Accepted: 08/01/2012] [Indexed: 11/27/2022]
Abstract
Deep hypothermia protects the brain from ischemic damage and is therefore used during major cardiovascular surgeries requiring cardiopulmonary bypass and a period of circulatory arrest. Here, we demonstrated that small ubiquitin-like modifier (SUMO1-3) conjugation is markedly activated in the brain during deep to moderate hypothermia. Animals were subjected to normothermic (37°C) or deep to moderate (18°C, 24°C, 30°C) hypothermic cardiopulmonary bypass, and the effects of hypothermia on SUMO conjugation were evaluated by Western blot and immunohistochemistry. Exposure to moderate 30°C hypothermia was sufficient to markedly increase levels and nuclear accumulation of SUMO2/3-conjugated proteins in these cells. Deep hypothermia induced nuclear translocation of the SUMO-conjugating enzyme Ubc9, suggesting that the increase in nuclear levels of SUMO2/3-conjugated proteins observed in brains of hypothermic animals is an active process. Exposure of primary neuronal cultures to deep hypothermia induced only a moderate rise in levels of SUMO2/3-conjugated proteins. This suggests that neurons in vivo have a higher capacity than neurons in vitro to activate this endogenous potentially neuroprotective pathway upon exposure to hypothermia. Identifying proteins that are SUMO2/3 conjugated during hypothermia could help to design new strategies for preventive and therapeutic interventions to make neurons more resistant to a transient interruption of blood supply.
Collapse
Affiliation(s)
- Liangli Wang
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
26
|
Kitagawa K. Ischemic tolerance in the brain: endogenous adaptive machinery against ischemic stress. J Neurosci Res 2012; 90:1043-54. [PMID: 22302606 DOI: 10.1002/jnr.23005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 10/25/2011] [Accepted: 11/18/2011] [Indexed: 01/10/2023]
Abstract
Although more than 100 drugs have been examined clinically, tissue plasminogen activator remains the only drug approved for the treatment of acute ischemic stroke. Since the discovery of ischemic tolerance, it has been widely recognized that the brain possesses an endogenous protective machinery to protect against ischemic stress. Recent studies have clarified that both the upregulation of neuroprotective signaling and the downregulation of inflammatory or apoptotic pathways are involved equally in the acquisition of ischemic tolerance. The triggering stimuli for ischemic stresses are divided into hypoxic, oxidant/inflammatory, and glutamate stress. Glutamate stress, particularly the synaptic stimulation of the N-methyl-D-aspartate receptor, leads to activation of the cAMP response element-binding protein, which could subsequently induce gene expression of several neuroprotective molecules. Gene reprogramming and metabolic downregulation are intimately involved in ischemic tolerance as well as in hibernation and hypothermia. Micro-RNAs may be a key player for tuning the level of gene expression in ischemic tolerance. Future research should be performed to investigate the most effective combination for brain protection, enhancement of cell survival signaling, and inhibition of the inflammatory or apoptotic pathways.
Collapse
Affiliation(s)
- Kazuo Kitagawa
- Department of Neurology, Stroke Center, Osaka University Graduate School of Medicine, Suita, Japan.
| |
Collapse
|
27
|
Hypoxic-preconditioning induces neuroprotection against hypoxia–ischemia in newborn piglet brain. Neurobiol Dis 2011; 43:473-85. [DOI: 10.1016/j.nbd.2011.04.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Revised: 04/15/2011] [Accepted: 04/22/2011] [Indexed: 11/20/2022] Open
|
28
|
Mitchell HM, White DM, Domowicz MS, Kraig RP. Cold pre-conditioning neuroprotection depends on TNF-α and is enhanced by blockade of interleukin-11. J Neurochem 2011; 117:187-96. [PMID: 21070241 PMCID: PMC3635118 DOI: 10.1111/j.1471-4159.2010.07103.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cold pre-conditioning reduces subsequent brain injury in small animals but the underlying mechanisms remain undefined. As hypothermia triggers systemic macrophage tumor necrosis factor alpha (TNF-α) production and other neural pre-conditioning stimuli depend on this cytokine, we reasoned that microglia and TNF-α would be similarly involved with cold pre-conditioning neuroprotection. Also, as slice cultures closely approximate their in vivo counterpart and include quiescent microglia, we used rat hippocampal slice cultures to confirm this hypothesis. Furthermore, inflammatory cytokine gene screening with subsequent PCR and immunostaining confirmation of targeted mRNA and related protein changes showed that cold pre-conditioning triggered a significant rise in TNF-α that localized to microglia and a significant rise in interleukin (IL)-11 that localized mainly to hippocampal pyramidal neurons and, more rarely, astrocytes. Importantly, co-stimulation with cold and IL-11, an anti-inflammatory cytokine that inhibits TNF-α expression, abrogated the otherwise evident protection. Instead, cold pre-conditioning coupled with blockade of IL-11 signaling further enhanced neuroprotection from that seen with cold pre-conditioning alone. Thus, physiological activation of brain pro-inflammatory cytokine signaling, and its amplification by inhibition of coincident anti-inflammatory cytokine signaling, may be opportune targets for the development of novel therapeutics that can mimic the protection seen in cold pre-conditioning.
Collapse
Affiliation(s)
- Heidi M. Mitchell
- Department of Neurology, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - David M. White
- Department of Neurology, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Miriam S. Domowicz
- Department of Pediatrics, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Richard P. Kraig
- Department of Neurology, The University of Chicago Medical Center, Chicago, Illinois, USA
| |
Collapse
|
29
|
Head BP, Peart JN, Panneerselvam M, Yokoyama T, Pearn ML, Niesman IR, Bonds JA, Schilling JM, Miyanohara A, Headrick J, Ali SS, Roth DM, Patel PM, Patel HH. Loss of caveolin-1 accelerates neurodegeneration and aging. PLoS One 2010; 5:e15697. [PMID: 21203469 PMCID: PMC3009734 DOI: 10.1371/journal.pone.0015697] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 11/29/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The aged brain exhibits a loss in gray matter and a decrease in spines and synaptic densities that may represent a sequela for neurodegenerative diseases such as Alzheimer's. Membrane/lipid rafts (MLR), discrete regions of the plasmalemma enriched in cholesterol, glycosphingolipids, and sphingomyelin, are essential for the development and stabilization of synapses. Caveolin-1 (Cav-1), a cholesterol binding protein organizes synaptic signaling components within MLR. It is unknown whether loss of synapses is dependent on an age-related loss of Cav-1 expression and whether this has implications for neurodegenerative diseases such as Alzheimer's disease. METHODOLOGY/PRINCIPAL FINDINGS We analyzed brains from young (Yg, 3-6 months), middle age (Md, 12 months), aged (Ag, >18 months), and young Cav-1 KO mice and show that localization of PSD-95, NR2A, NR2B, TrkBR, AMPAR, and Cav-1 to MLR is decreased in aged hippocampi. Young Cav-1 KO mice showed signs of premature neuronal aging and degeneration. Hippocampi synaptosomes from Cav-1 KO mice showed reduced PSD-95, NR2A, NR2B, and Cav-1, an inability to be protected against cerebral ischemia-reperfusion injury compared to young WT mice, increased Aβ, P-Tau, and astrogliosis, decreased cerebrovascular volume compared to young WT mice. As with aged hippocampi, Cav-1 KO brains showed significantly reduced synapses. Neuron-targeted re-expression of Cav-1 in Cav-1 KO neurons in vitro decreased Aβ expression. CONCLUSIONS Therefore, Cav-1 represents a novel control point for healthy neuronal aging and loss of Cav-1 represents a non-mutational model for Alzheimer's disease.
Collapse
Affiliation(s)
- Brian P. Head
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- VA San Diego Healthcare System, San Diego, California, United States of America
| | - Jason N. Peart
- Heart Foundation Research Centre, Griffith University, Gold Coast, Queensland, Australia
| | - Mathivadhani Panneerselvam
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Takaakira Yokoyama
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Matthew L. Pearn
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Ingrid R. Niesman
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Jacqueline A. Bonds
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Jan M. Schilling
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- VA San Diego Healthcare System, San Diego, California, United States of America
| | - Atsushi Miyanohara
- Gene Therapy Program, University of California San Diego, La Jolla, California, United States of America
| | - John Headrick
- Heart Foundation Research Centre, Griffith University, Gold Coast, Queensland, Australia
| | - Sameh S. Ali
- Department of Medicine, University of California, La Jolla, California, United States of America
| | - David M. Roth
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- VA San Diego Healthcare System, San Diego, California, United States of America
| | - Piyush M. Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- VA San Diego Healthcare System, San Diego, California, United States of America
| | - Hemal H. Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- VA San Diego Healthcare System, San Diego, California, United States of America
| |
Collapse
|
30
|
Kraig RP, Mitchell HM, Christie-Pope B, Kunkler PE, White DM, Tang YP, Langan G. TNF-α and Microglial Hormetic Involvement in Neurological Health & Migraine. Dose Response 2010; 8:389-413. [PMID: 21191481 DOI: 10.2203/dose-response.09-056.kraig] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Environmental enrichment, i.e., increased intellectual, social, and physical activity makes brain more resilient to subsequent neurological disease. The mechanisms for this effect remain incompletely defined, but evidence shows tumor necrosis factor-alpha (TNF-α) is involved. TNF-α, at acutely high levels, possesses the intrinsic capacity to enhance injury associated with neurological disease. Conversely, the effect of TNF-α at low-levels is nutritive over time, consistent with physiological conditioning hormesis. Evidence shows that neural activity triggers low-level pro-inflammatory signaling involving TNF-α. This low-level TNF-α signaling alters gene expression, resulting in an enhanced resilience to disease. Brain-immune signaling may become maladaptive when increased activity is chronic without sufficient periods of reduced activity necessary for nutritive adaptation. Such tonically increased activity may explain, for example, the transformation of episodic to chronic migraine with related increased susceptibility to spreading depression, the most likely underlying cause of this malady. Thus, TNF-α, whose function is to alter gene expression, and its principal cellular source, microglia, seem powerfully positioned to orchestrate hormetic immune signaling that establishes the phenotype of neurological health and disease from brain activity.
Collapse
Affiliation(s)
- Richard P Kraig
- Department of Neurology, The University of Chicago Medical Center, Chicago, IL
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Preconditioning (PC) describes a phenomenon whereby a sub-injury inducing stress can protect against a later injurious stress. Great strides have been made in identifying the mechanisms of PC-induced protection in animal models of brain injury. While these may help elucidate potential therapeutic targets, there are questions over the clinical utility of cerebral PC, primarily because of questions over the need to give the PC stimulus prior to the injury, narrow therapeutic windows and safety. The object of this review is to address the question of whether there may indeed be a clinical use for cerebral PC and to discuss the deficiencies in our knowledge of PC that may hamper such clinical translation.
Collapse
Affiliation(s)
- Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael M. Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jianming Xiang
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
|
33
|
Mechírová E, Domoráková I, Danková M, Danielisová V, Burda J. Effect of noradrenalin and EGb 761 pretreatment on the ischemia-reperfusion injured spinal cord neurons in rabbits. Cell Mol Neurobiol 2009; 29:991-8. [PMID: 19291391 PMCID: PMC11505823 DOI: 10.1007/s10571-009-9386-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Accepted: 02/27/2009] [Indexed: 01/07/2023]
Abstract
Short term sublethal ischemia or ischemic preconditioning gives protection to the neurons against subsequent lethal ischemic attack. This so-called ischemic tolerance can also be provided by certain drugs. We examined the effect of noradrenalin and EGb 761 on the spinal cord neurons injured by 30 min occlusion of abdominal aorta in rabbits. The animals survived 48 and 72 h. Degenerated neurons were visualized by Fluoro Jade B method, viable neurons were demonstrated immunohistochemically with NeuN and ubiquitin antibodies. The rabbits with noradrenalin administration 48 h before 30 min of ischemia and 48/72 h of reperfusion, showed significant increase of degenerated Fluoro Jade B labeled neurons. Animals of both groups were paraplegic. Rabbits pretreated 7 days with EGb 761 prior to 30 min of ischemia and with 48/72 h of reperfusion revealed significant decrease of Fluoro Jade B-positive neurons when compared with the groups with 30 min of ischemia followed by 48/72 h of reperfusion. In the NeuN sections, the number of viable neurons was moderately decreased. These animals showed no paraplegia. Ubiquitin aggregates occurred in the cytoplasm of degenerated neurons in the sections of rabbits preconditioned with noradrenalin 48 h prior to 30 min of ischemia and followed by 48 h of reperfusion while after 72 h of reperfusion, shrunk light shadows without ubiquitin reaction were visible. Our results indicate that EGb 761 could be involved in protection of spinal cord neurons against ischemic injury while effect of noradrenalin is not unambiguous.
Collapse
Affiliation(s)
- Eva Mechírová
- Department of Histology and Embryology, Faculty of Medicine, PJ Safárik University in Kosice, 04180 Kosice, Slovak Republic.
| | | | | | | | | |
Collapse
|
34
|
Benardete EA, Bergold PJ. Genomic analysis of ischemic preconditioning in adult rat hippocampal slice cultures. Brain Res 2009; 1292:107-22. [PMID: 19631194 DOI: 10.1016/j.brainres.2009.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Revised: 07/04/2009] [Accepted: 07/08/2009] [Indexed: 01/08/2023]
Abstract
Understanding endogenous mechanisms of neuroprotection may have important clinical applications. It is well established that brain tissue becomes more resistant to ischemic injury following a sublethal ischemic insult. This process, called ischemic preconditioning (IPC), can be induced in adult rat hippocampal slice cultures by a brief oxygen-glucose deprivation (OGD) [Hassen, G.W., Tian, D., Ding, D., Bergold, P.J., 2004. A new model of ischemic preconditioning using young adult hippocampal slice cultures. Brain Res. Brain Res. Protoc. 13, 135-143]. We have analyzed the changes in gene expression brought about by IPC in this model in order to understand the mechanisms involved. Total RNA was isolated at different time points following a brief OGD (3, 6 and 12 h) and used to probe genome-wide expression microarrays. Genes were identified that were significantly up- or down-regulated relative to controls. We placed genes that were differentially expressed into statistically significant groups based on Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and gene ontology (GO) terms. Genes involved in signal transduction, transcription, and oxidative phosphorylation are differentially expressed at each time point. The analysis demonstrates that alterations in signaling pathways (TGF-beta, Wnt, MAPK, ErbB, Toll-like receptor, JAK-STAT, VEGF) consistently accompany IPC. RT-PCR was used to confirm that members of these signaling pathways are regulated as predicted by the microarray analysis. We verified that protein translation following OGD is necessary for IPC. We also found that blocking the NMDA receptor during OGD does not significantly inhibit IPC in this model or produce large changes in gene expression. Our data thus suggests that changes in signaling pathways and their down-stream targets play an important role in triggering endogenous neuroprotection.
Collapse
Affiliation(s)
- Ethan A Benardete
- Department of Neurosurgery, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | |
Collapse
|
35
|
Neuronal plasticity after ischemic preconditioning and TIA-like preconditioning ischemic periods. Acta Neuropathol 2009; 117:511-23. [PMID: 19084975 DOI: 10.1007/s00401-008-0473-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 11/29/2008] [Accepted: 12/06/2008] [Indexed: 01/10/2023]
Abstract
Transient ischemic attacks (TIAs) have recently become the center of attention since they are thought to share some characteristics with experimental ischemic preconditioning (IPC). This phenomenon describes the situation that a brief, per se harmless, cerebral ischemic period renders the brain resistant to a subsequent severe and normally damaging ischemia. Preconditioning (PC) is not restricted to the brain but also occurs in other organs. Furthermore, apart from a short ischemia, the PC event may comprise nearly any noxious stimulus which, however, must not exceed the threshold to tissue damage. In the last two decades, our knowledge concerning the underlying molecular basis of PC has substantially grown and there is hope to potentially imitate the induction of an endogenous neuroprotective state in patients with a high risk of cerebral ischemia. While, at present, there is virtually no neuropathological data on changes after TIAs or TIA-like PC ischemic periods in human brains, the following review will briefly summarize the current knowledge of plastic neuronal changes after PC in animal models, still awaiting their detection in the human brain.
Collapse
|
36
|
Yang W, Ma Q, Mackensen GB, Paschen W. Deep hypothermia markedly activates the small ubiquitin-like modifier conjugation pathway; implications for the fate of cells exposed to transient deep hypothermic cardiopulmonary bypass. J Cereb Blood Flow Metab 2009; 29:886-90. [PMID: 19240742 DOI: 10.1038/jcbfm.2009.16] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Various cardiovascular operations are performed during conditions of deep hypothermic circulatory arrest. Here we investigated the effects of deep hypothermia on the small ubiquitin-like modifier (SUMO) conjugation pathway using a clinically relevant animal model of deep hypothermic cardiopulmonary bypass (DHCPB). Deep hypothermic cardiopulmonary bypass induced a marked activation of the SUMO conjugation pathway and triggered a nuclear translocation of SUMO2/3-conjugated proteins. Furthermore, DHCBP significantly modified gene expression. Activation of the SUMO conjugation pathway is believed to protect neurons from damage caused by low blood flow. This pathway may, therefore, play a key role in defining the outcome of cells exposed to DHCPB.
Collapse
Affiliation(s)
- Wei Yang
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
37
|
Katz LM, Frank JE, Dvorak A, Finch A, Szymanowski A, Gordon CJ. Independence of brain and trunk temperature during hypothermic preconditioning in rats. J Neurosci Methods 2009; 179:179-83. [PMID: 19428525 DOI: 10.1016/j.jneumeth.2009.01.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 01/24/2009] [Accepted: 01/26/2009] [Indexed: 12/01/2022]
Abstract
UNLABELLED Hypothermic preconditioning is rapid cooling and warming to induce tolerance to ischemia. The purpose of the study was to examine differences in brain and trunk temperature during hypothermic preconditioning. METHODS Rats (n=18) were implanted with telemetric probes for simultaneous measure of brain and trunk temperature. Hypothermic preconditioning was produced by exposing rats to cool and warm environments that produced rapid cooling to 30 degrees C and warming to 35 degrees C. RESULTS Brain temperature was warmer (37.56+/-0.45 degrees C) than trunk (37.17+/-0.29 degrees C) temperature in unanesthetized, free roaming rats at room temperature (t-test p=0.04). The brain cooled (0.59+/-0.1 degrees C/min) quicker than the trunk (0.44+/-0.19 degrees C/min) during cooling cycles of hypothermic preconditioning and the brain (0.28+/-0.04 degrees C/min) warmed quicker than the trunk (0.18+/-0.07 degrees C/min) during the warming cycle of hypothermic preconditioning (t-test p<0.0001). When the trunk temperature probe was designated to reach the target temperature of 35 degrees C during warming, the brain temperature (38.1+/-0.44 degrees C) was warmer than trunk temperature (34.95+/-0.16 degrees C) during the peak of warming (t-test p<0.0001). CONCLUSION The brain cools and warms quicker than the trunk during hypothermic preconditioning. Failure to anticipate these differences could lead to unrecognized brain hyperthermia during warming. Appreciation of differences in rates of change between brain and trunk temperature may be important when designing hypothermic preconditioning experiments.
Collapse
Affiliation(s)
- Laurence M Katz
- University of North Carolina School of Medicine, Department of Emergency Medicine, CB 7594, Neurosciences Hospital Ground Floor, 101 Manning Drive, Chapel Hill, NC 27599, USA.
| | | | | | | | | | | |
Collapse
|
38
|
STETLER RANNE, ZHANG FENG, LIU COLLIN, CHEN JUN. Ischemic tolerance as an active and intrinsic neuroprotective mechanism. HANDBOOK OF CLINICAL NEUROLOGY 2009; 92:171-95. [PMID: 18790275 PMCID: PMC2710312 DOI: 10.1016/s0072-9752(08)01909-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - FENG ZHANG
- University of Pittsburgh, Pittsburgh, PA, USA
| | - COLLIN LIU
- University of Pittsburgh, Pittsburgh, PA, USA
| | - JUN CHEN
- University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
39
|
Zecher M, Guichard C, Velásquez MJ, Figueroa G, Rodrigo R. Implications of oxidative stress in the pathophysiology of obstructive uropathy. ACTA ACUST UNITED AC 2008; 37:19-26. [PMID: 19082822 DOI: 10.1007/s00240-008-0163-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 11/25/2008] [Indexed: 02/07/2023]
Abstract
Although the functional and clinical alterations occurring in patients with obstructive uropathy are not well understood, it has been suggested that oxidative stress could contribute in the mechanism responsible for the impairment of sodium and water balance. This study aimed to test the hypothesis that red wine administration causes an amelioration of both the renal damage and impairment of renal Na(+), K(+)-ATPase activity occurring after ureteral obstruction in the rat. Twenty-four male Wistar adult rats weighting 200-250 g were used. Half of them received a 10-week treatment with wine as the sole fluid source, while the other group received water. Both groups were subjected to 24-h unilateral ureteral obstruction (UUO). Kidney tissue was collected following the relief of the ligature to perform the biochemical assessments. Urine and blood samples were taken at baseline and after the relief. Results show that the treatment with red wine significantly enhances the activity of antioxidant enzymes, and thus reduces renal lipid peroxidation secondary to UUO, which correlated negatively with Na(+), K(+)-ATPase activity. Based on this and other previous data, it could be suggested that red wine administration may prevent renal damage secondary to UUO by inducing enhanced antioxidant potential.
Collapse
Affiliation(s)
- Martin Zecher
- Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
40
|
Yenari M, Kitagawa K, Lyden P, Perez-Pinzon M. Metabolic downregulation: a key to successful neuroprotection? Stroke 2008; 39:2910-7. [PMID: 18658035 PMCID: PMC2597567 DOI: 10.1161/strokeaha.108.514471] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Accepted: 04/02/2008] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND PURPOSE The search for effective neuroprotectants remains frustrating, particularly with regard to specific pharmaceuticals. However, laboratory studies have consistently shown remarkable neuroprotection with 2 nonpharmacological strategies-therapeutic hypothermia and ischemic preconditioning. Recent studies have shown that the mechanism of protection underlying both of these treatments is correlated to downregulation of cellular and tissue metabolism. Thus, understanding the mechanisms underlying such robust protective effects could lead to appropriate translation at the clinical level. In fact, hypothermia is already being used at many centers to improve neurological outcome from cardiac arrest. METHODS A systematic review of both topics is presented in terms of underlying pathophysiological mechanisms and application at the clinical level. RESULTS Although the mechanisms of protection for both therapeutic strategies are multifold, both share features of downregulating metabolism. Both therapeutic strategies are robust neuroprotectants, but translating them to the clinical arena is challenging, though not impossible, and clinical studies have shown or suggest benefits of both treatments. CONCLUSIONS The strategy of metabolic downregulation should be further explored to identify effective neuroprotectants that can be easily applied clinically.
Collapse
Affiliation(s)
- Midori Yenari
- Department of Neurology, University of California, San Francisco, San Francisco Veterans Affairs Medical Center, San , San Francisco, CA 94121, USA.
| | | | | | | |
Collapse
|
41
|
Christian SL, Ross AP, Zhao HW, Kristenson HJ, Zhan X, Rasley BT, Bickler PE, Drew KL. Arctic ground squirrel (Spermophilus parryii) hippocampal neurons tolerate prolonged oxygen-glucose deprivation and maintain baseline ERK1/2 and JNK activation despite drastic ATP loss. J Cereb Blood Flow Metab 2008; 28:1307-19. [PMID: 18398417 PMCID: PMC2792705 DOI: 10.1038/jcbfm.2008.20] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Oxygen-glucose deprivation (OGD) initiates a cascade of intracellular responses that culminates in cell death in sensitive species. Neurons from Arctic ground squirrels (AGS), a hibernating species, tolerate OGD in vitro and global ischemia in vivo independent of temperature or torpor. Regulation of energy stores and activation of mitogen-activated protein kinase (MAPK) signaling pathways can regulate neuronal survival. We used acute hippocampal slices to investigate the role of ATP stores and extracellular signal-regulated kinase (ERK)1/2 and Jun NH(2)-terminal kinase (JNK) MAPKs in promoting survival. Acute hippocampal slices from AGS tolerated 30 mins of OGD and showed a small but significant increase in cell death with 2 h OGD at 37 degrees C. This tolerance is independent of hibernation state or season. Neurons from AGS survive OGD despite rapid ATP depletion by 3 mins in interbout euthermic AGS and 10 mins in hibernating AGS. Oxygen-glucose deprivation does not induce JNK activation in AGS and baseline ERK1/2 and JNK activation is maintained even after drastic depletion of ATP. Surprisingly, inhibition of ERK1/2 or JNK during OGD had no effect on survival, whereas inhibition of JNK increased cell death during normoxia. Thus, protective mechanisms promoting tolerance to OGD by AGS are downstream from ATP loss and are independent of hibernation state or season. Journal of Cerebral Blood Flow & Metabolism (2008) 28, 1307-1319; doi:10.1038/jcbfm.2008.20; published online 9 April 2008.
Collapse
Affiliation(s)
- Sherri L Christian
- Alaskan Basic Neuroscience Program, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, Alaska 99775-7000, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Ischemic brain damage can be prevented or at least significantly reduced when there is a preceding brief ischemic period that does not exceed the threshold for tissue damage--a phenomenon termed "ischemic preconditioning" (ischemic PC). Experimental PC in rodents is now considered to be a model for transient ischemic attacks in humans, and there is increasing hope for translating the knowledge of underlying mechanisms in the animal models into the clinic to enhance endogenous neuroprotective mechanisms in patients with stroke. However, although PC was originally defined as a subtoxic stimulus without any morphologic damage, there is a growing body of evidence from studies using sensitive techniques that postischemic structural alterations of brain tissue manifest not only after ischemia with prior PC but also after the PC stimulus itself. Furthermore, it has become evident over time that the primary shortcomings of many experimental studies on PC are the short observation intervals. The few studies with extended postischemic survival periods done to date provide clear evidence of considerable structural changes and even cell death, which may only be postponed by PC. Therefore, further studies are needed to elucidate structural long-term changes after PC and to validate the persistence of the neuroprotective effects.
Collapse
|
43
|
Zhang N, Gao G, Bu X, Han S, Fang L, Li J. Neuron-specific phosphorylation of c-Jun N-terminal kinase increased in the brain of hypoxic preconditioned mice. Neurosci Lett 2007; 423:219-24. [PMID: 17709198 DOI: 10.1016/j.neulet.2007.07.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Revised: 07/04/2007] [Accepted: 07/06/2007] [Indexed: 10/23/2022]
Abstract
Accumulated studies have suggested that mitogen-activated protein kinase (MAPK) play a pivotal role in the development of cerebral hypoxic preconditioning (HPC). By using our "auto-hypoxia"-induced HPC mouse model, we have reported increased phosphorylation level of p38 MAPK, and decreased phosphorylation and protein expression levels of extracellular signal regulated kinases 1/2 (ERK1/2) in the brain of HPC mice. In the current study, we investigated the involvement of c-Jun N-terminal kinase (JNK) in the brain of HPC mice. By using Western blot analysis, we found that the phosphorylation levels of JNK at Thr183 and Tyr185 sites (phospho-Thr183/Tyr185 JNK), but not its protein expression, increased significantly (p<0.05, n=6 for each group) both in the hippocampus and frontal cortex of early (H1-H4) and delayed (H5 and H6) HPC mice than that of the normoxic group (H0, n=6). Similarly, enhanced phospho-Thr183/Tyr185 JNK was also observed by immunostaining in the hippocampus and frontal cortex of mice following series of hypoxic exposures (H3 and H6). In addition, we found that phospho-Thr183/Tyr185 JNK predominantly co-localized with a neuron-specific protein, neurogranin, in both the hippocampus and frontal cortex of HPC mice (H3) by using double-labeled immunofluorescence. These results suggest that the increased neuron-specific phosphorylation of JNK at Thr183/Tyr185, not protein expression, might be involved in the development of cerebral HPC of mice.
Collapse
Affiliation(s)
- Nan Zhang
- Institute for Biomedical Science of Pain, Beijing Key Laboratory for Neural Regeneration and Repairing, Department of Neurobiology, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing 100069, China
| | | | | | | | | | | |
Collapse
|
44
|
Qi Z, Bu X, Huang P, Zhang N, Han S, Fang L, Li J. Increased Membrane/Nuclear Translocation and Phosphorylation of p90 KD Ribosomal S6 Kinase in the Brain of Hypoxic Preconditioned Mice. Neurochem Res 2007; 32:1450-9. [PMID: 17404833 DOI: 10.1007/s11064-007-9331-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Accepted: 03/07/2007] [Indexed: 10/23/2022]
Abstract
Our previous studies have demonstrated that hypoxic precondition (HPC) increased membrane translocation of protein kinase C isoforms and decreased phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) in the brain of mice. The goal of this study was to determine the involvement of p90 KD ribosomal S6 kinase (RSK) in cerebral HPC of mice. Using Western-blot analysis, we found that the levels of membrane/nuclear translocation, but not protein expression of RSK increased significantly in the frontal cortex and hippocampus of HPC mice. In addition, we found that the phosphorylation levels of RSK at the Ser227 site (a PDK1 phosphorylation site), but not at the Thr359/Ser363 sites (ERK1/2 phosphorylated sites) increased significantly in the brain of HPC mice. Similar results were confirmed by an immunostaining study of total RSK and phospho-Ser227 RSK. To further define the cellular populations to express phospho-Ser227 RSK, we found that the expression of phospho-Ser227 RSK co-localized with neurogranin, a neuron-specific marker, in cortex and hippocampus of HPC mice by using double-labeled immunofluorescent staining method. These results suggest that increased RSK membrane/nuclear translocation and PDK1 mediated neuron-specific phosphorylation of RSK at Ser227 might be involved in the development of cerebral HPC of mice.
Collapse
Affiliation(s)
- Zhifeng Qi
- Department of Neurobiology, Beijing Key Laboratory for Neural Regeneration and Repairing, Institute for Biomedical Science of Pain, Capital Medical University, #10 You An Men Wai Xi Tou Tiao, Beijing 100069, Peoples' Republic of China
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Many stressful, but not lethal, stimuli activate endogenous protective mechanisms that significantly decrease the degree of injury to subsequent injurious stimuli. This protective mechanism is termed preconditioning and tolerance. It occurs across organ systems including the brain and nervous system. Preconditioning has been investigated in cell and animal models and recently been shown to potentially occur in human brain. Learning more about these powerful endogenous neuroprotective mechanisms could help identify new approaches to treat patients with stroke and other central nervous system disorders or injury. Cell and animal models are helping us to better understand the network response of gene and protein expression that activates the neuroprotective response.
Collapse
Affiliation(s)
- V L Dawson
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, USA.
| | | |
Collapse
|
46
|
Akaiwa K, Akashi H, Harada H, Sakashita H, Hiromatsu S, Kano T, Aoyagi S. Moderate cerebral venous congestion induces rapid cerebral protection via adenosine A1 receptor activation. Brain Res 2006; 1122:47-55. [PMID: 17067559 DOI: 10.1016/j.brainres.2006.09.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Revised: 08/31/2006] [Accepted: 09/03/2006] [Indexed: 11/25/2022]
Abstract
Stroke is a devastating complication in cardiovascular surgery, and neuronal damage is worsened by intracranial pressure elevation caused by cerebral venous circulatory disturbances (CVCD). However, we have previously reported that CVCD before cerebral ischemia decreases the infarct area. In the present study, focal cerebral ischemia was induced in spontaneously hypertensive rats by filament insertion through the carotid artery. Rats were divided into the following four groups: sham-operated, mild or severe venous congestion (VC), and DPCPX. The DPCPX group received the adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) prior to mild VC. Behavior, infarct volume, edema and S-100 protein were evaluated among the four groups. The infarct volume rates in mild VC and severe VC groups were significantly less than that in sham-operated and DPCPX groups. However, the mortality of the severe VC group worsened in a time-dependent manner. We observed a significant decrease in edema in the mild VC group compared to the DPCPX group. Behavioral scores also indicated that the mild VC group had fewer neurological deficits than the other three groups, including the DPCPX group. We were able to induce rapid cerebral protection via adenosine A1 receptor activation by administering an appropriate degree of VC prior to cerebral ischemia produced by middle cerebral artery occlusion. Our work suggests possible mechanisms by which such effective VC may lead to cerebral protection and adenosine A1 receptor activation.
Collapse
Affiliation(s)
- Keiichi Akaiwa
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Burda J, Danielisová V, Némethová M, Gottlieb M, Matiasová M, Domoráková I, Mechírová E, Feriková M, Salinas M, Burda R. Delayed postconditionig initiates additive mechanism necessary for survival of selectively vulnerable neurons after transient ischemia in rat brain. Cell Mol Neurobiol 2006; 26:1141-51. [PMID: 16612578 PMCID: PMC11520615 DOI: 10.1007/s10571-006-9036-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Accepted: 12/09/2005] [Indexed: 10/24/2022]
Abstract
1. The aim of this study was to validate the role of postconditioning, used 2 days after lethal ischemia, for protection of selectively vulnerable brain neurons against delayed neuronal death. 2. Eight, 10, or 15 min of transient forebrain ischemia in rat (four-vessel occlusion model) was used as initial lethal ischemia. Fluoro Jade B, the marker of neurodegeneration, and NeuN, a specific neuronal marker were used for visualization of changes 7 or 28 days after ischemia without and with delayed postconditioning. 3. Our results confirm that postconditioning if used at right time and with optimal intensity can prevent process of delayed neuronal death. At least three techniques, known as preconditioners, can be used as postconditioning: short ischemia, 3-nitropropionic acid and norepinephrine. A cardinal role for the prevention of death in selectively vulnerable neurons comprises synthesis of proteins during the first 5 h after postconditioning. Ten minutes of ischemia alone is lethal for 70% of pyramidal CA1 neurons in hippocampus. Injection of inhibitor of protein synthesis (Cycloheximide), if administered simultaneously with postconditioning, suppressed beneficial effect of postconditioning and resulted in 50% of CA1 neurons succumbing to neurodegeneration. Although, when Cycloheximide was injected 5 h after postconditioning, this treatment resulted in survival of 90% of CA1 neurons. 4. Though postconditioning significantly protects hippocampal CA1 neurons up to 10 min of ischemia, its efficacy at 15 min ischemia is exhausted. However, protective impact of postconditioning in less-sensitive neuronal populations (cortex and striatum) is very good after such a damaging insult like 15 min ischemia. This statement also means that up to 15 min of ischemia, postconditioning does not induce cumulation of injuries produced by the first and the second stress.
Collapse
Affiliation(s)
- Jozef Burda
- Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovakia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gurcun U, Discigil B, Boga M, Ozkisacik E, Badak MI, Yenisey C, Kurtoglu T, Meteoglu I. Is remote preconditioning as effective as direct ischemic preconditioning in preventing spinal cord ischemic injury? J Surg Res 2006; 135:385-93. [PMID: 16904694 DOI: 10.1016/j.jss.2006.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Revised: 03/31/2006] [Accepted: 04/03/2006] [Indexed: 10/24/2022]
Abstract
BACKGROUND Spinal cord injury remains a devastating complication of thoracic and thoracoabdominal aortic operations. The aim of this study was to assess the affectivity of direct ischemic preconditioning (PC) and remote PC in preventing spinal cord ischemic injury in an experimental model. MATERIALS AND METHODS Thirty-eight New Zealand white rabbits were divided into five groups: One group served as Sham group (n = 7). Rabbits in other groups had their abdominal aorta cross-clamped for 40 min. Before aortic occlusion, aorta was clamped twice at the same site of aortic occlusion for 5 min followed by 15 min of reperfusion after each ischemic episode in one group (Direct PC, n = 8), left renal artery was clamped twice for 5 min followed by 15 min of reperfusion after each renal ischemic episode in one group (Remote PC, n = 8), left renal artery was first clamped for 5 min followed by 15 min of reperfusion and then aorta was clamped for 5 min followed by 15 min of reperfusion in one group (Remote + Direct PC, n = 8), and no PC method was used in Control group (n = 7). RESULTS In all PC groups, neurological status of rabbits (Tarlov score) at post-ischemia 24th and 48th hours was better than the control group (P < 0.05), but worse than Sham group (P < 0.05). Mean viability index values in PC groups were higher than control group (P < 0.01). Post-ischemia serum NSE and MDA levels obtained in all three PC groups were significantly lower than control group (P < 0.05 and P < 0.01). CONCLUSIONS The use of direct ischemic PC and/or remote PC is an effective way of reducing spinal cord ischemic injury because of aortic occlusion, while direct PC is more effective. The combined use of direct PC and remote PC did not provide better protection.
Collapse
Affiliation(s)
- Ugur Gurcun
- Department of Cardiovascular Surgery, Adnan Menderes University Medical Faculty, Aydin, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Yuan HB, Huang Y, Zheng S, Zuo Z. Hypothermic preconditioning reduces Purkinje cell death possibly by preventing the over-expression of inducible nitric oxide synthase in rat cerebellar slices after an in vitro simulated ischemia. Neuroscience 2006; 142:381-9. [PMID: 16890370 DOI: 10.1016/j.neuroscience.2006.06.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Revised: 06/07/2006] [Accepted: 06/23/2006] [Indexed: 11/24/2022]
Abstract
We showed that hypothermic preconditioning (HPC) increased survival of Purkinje neurons in rat cerebellar slices after oxygen-glucose deprivation (OGD). HPC also reduced the OGD-increased expression of high mobility group I (Y) proteins, a transcription factor that can enhance inducible nitric oxide synthase (iNOS) expression. iNOS is a putatively damaging protein that contributes to ischemic brain injury. Heat shock proteins (HSPs) can be induced by various stimuli to protect cells. We hypothesize that HPC induces neuroprotection by reducing the expression of putatively damaging proteins such as iNOS and/or by increasing the expression of putatively protective proteins such as HSPs. Cerebellar slices were prepared from adult male Sprague-Dawley rats and incubated in circulating artificial cerebrospinal fluid. OGD was for 20 min at 37 degrees C and was followed by a 5-h recovery at 37 degrees C before slices were used for morphological, immunohistochemical and Western analyses. HPC was performed by incubating slices at 33 degrees C for 20 min at 1 h before the OGD. HPC and aminoguanidine, an iNOS inhibitor, prevented OGD-induced Purkinje cell death/injury. OGD increased the expression of iNOS and nitrosylated proteins. These increases were abolished by aminoguanidine and HPC. Interestingly, the expression of HSP70 was increased by OGD but not by HPC. Our results suggest that an increased iNOS expression contributes to the pathophysiology of OGD-induced Purkinje neuronal death in our model. Our results also suggest the involvement of inhibiting the expression of the putatively damaging iNOS proteins in the HPC-induced neuroprotection. HSP70 may not contribute to the HPC-induced neuroprotection.
Collapse
Affiliation(s)
- H-B Yuan
- Department of Anesthesiology, University of Virginia Health System, One Hospital Drive, PO Box 800710, Charlottesville, VA 22908-0710, USA
| | | | | | | |
Collapse
|
50
|
Delgado P, Sahuquillo J, Poca MA, Alvarez-Sabin J. Neuroprotection in malignant MCA infarction. Cerebrovasc Dis 2006; 21 Suppl 2:99-105. [PMID: 16651820 DOI: 10.1159/000091709] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Massive unilateral hemispheric infarction often develops progressive postischemic edema that leads to a malignant course of stroke with mortality of up to 80% with conventional medical therapies. Hypothermia and decompressive hemicraniectomy have shown neuroprotective effects in several animal models of focal transient and permanent MCA occlusion by reducing infarct size and improving neurological outcome. Our aim in this paper was to review the possible mechanisms of both therapies as well as the optimal time window and duration of application of each treatment in animal model and in human malignant MCA infarction reported in the literature.
Collapse
Affiliation(s)
- Pilar Delgado
- Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain.
| | | | | | | |
Collapse
|