1
|
Kawahara M, Kato-Negishi M, Tanaka KI. Dietary Trace Elements and the Pathogenesis of Neurodegenerative Diseases. Nutrients 2023; 15:2067. [PMID: 37432185 PMCID: PMC10180548 DOI: 10.3390/nu15092067] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 07/12/2023] Open
Abstract
Trace elements such as iron (Fe), zinc (Zn), copper (Cu), and manganese (Mn) are absorbed from food via the gastrointestinal tract, transported into the brain, and play central roles in normal brain functions. An excess of these trace elements often produces reactive oxygen species and damages the brain. Moreover, increasing evidence suggests that the dyshomeostasis of these metals is involved in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, prion diseases, and Lewy body diseases. The disease-related amyloidogenic proteins can regulate metal homeostasis at the synapses, and thus loss of the protective functions of these amyloidogenic proteins causes neurodegeneration. Meanwhile, metal-induced conformational changes of the amyloidogenic proteins contribute to enhancing their neurotoxicity. Moreover, excess Zn and Cu play central roles in the pathogenesis of vascular-type senile dementia. Here, we present an overview of the intake, absorption, and transport of four essential elements (Fe, Zn, Cu, Mn) and one non-essential element (aluminum: Al) in food and their connections with the pathogenesis of neurodegenerative diseases based on metal-protein, and metal-metal cross-talk.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| |
Collapse
|
2
|
Vucic S, Pavey N, Haidar M, Turner BJ, Kiernan MC. Cortical hyperexcitability: Diagnostic and pathogenic biomarker of ALS. Neurosci Lett 2021; 759:136039. [PMID: 34118310 DOI: 10.1016/j.neulet.2021.136039] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/04/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Cortical hyperexcitability is an early and intrinsic feature of both sporadic and familial forms of amyotrophic lateral sclerosis (ALS).. Importantly, cortical hyperexcitability appears to be associated with motor neuron degeneration, possibly via an anterograde glutamate-mediated excitotoxic process, thereby forming a pathogenic basis for ALS. The presence of cortical hyperexcitability in ALS patients may be readily determined by transcranial magnetic stimulation (TMS), a neurophysiological tool that provides a non-invasive and painless method for assessing cortical function. Utilising the threshold tracking TMS technique, cortical hyperexcitability has been established as a robust diagnostic biomarker that distinguished ALS from mimicking disorders at early stages of the disease process. The present review discusses the pathophysiological and diagnostic utility of cortical hyperexcitability in ALS.
Collapse
Affiliation(s)
- Steve Vucic
- Western Clinical School, University of Sydney, Sydney, Australia.
| | - Nathan Pavey
- Western Clinical School, University of Sydney, Sydney, Australia
| | - Mouna Haidar
- Florey Institute of Neuroscieace and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscieace and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney and Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
3
|
Perioperative Pregabalin for Preventive Analgesia in Breast Cancer Surgery: A Meta-analysis of Randomized Controlled Trials. Clin J Pain 2020; 36:968-977. [PMID: 32960823 DOI: 10.1097/ajp.0000000000000883] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES Pregabalin is a drug for neuropathic pain. Antipronociceptive properties of pregabalin have led to its recent use as an adjuvant to the multimodal postoperative pain regimen. This meta-analysis was conducted to evaluate the efficacy of perioperative pregabalin on acute and chronic postsurgical pain (CPSP) after breast cancer surgery. METHODS A meta-analysis including 8 randomized controlled trials searched from MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials was conducted. Subgroup analysis was performed according to doses and timecourse of pregabalin administration. Review Manager 5.3 was selected to conduct the meta-analysis. RESULTS Preoperative pregabalin in breast cancer surgery alleviated acute postoperative pain at rest 24 hours after surgery by 0.31 points on an 0 to 10 Numerical Rating Scale (95% confidence interval [CI] -0.57 to -0.05). Morphine consumption showed a decrease in postoperative use by 1.09 mg (95% CI: -1.61 to -0.57). The incidence of CPSP 3 months after surgery was reduced to 46% (95% CI: 0.25-0.85). Postoperative nausea and vomiting, dizziness, and sedation showed no overall significant reductions. However, a decrease in the incidence of postoperative nausea and vomiting and an increase in the incidence of dizziness were noted when patients received 300 mg of pregabalin before surgery. DISCUSSION This study demonstrated that pregabalin showed more efficacy on chronic pain than acute pain after a breast cancer surgery. Further study based on doses and treatment course of pregabalin should be conducted to establish stronger evidence of treatment effects.
Collapse
|
4
|
Chandrasekaran K, Choi J, Arvas MI, Salimian M, Singh S, Xu S, Gullapalli RP, Kristian T, Russell JW. Nicotinamide Mononucleotide Administration Prevents Experimental Diabetes-Induced Cognitive Impairment and Loss of Hippocampal Neurons. Int J Mol Sci 2020; 21:ijms21113756. [PMID: 32466541 PMCID: PMC7313029 DOI: 10.3390/ijms21113756] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetes predisposes to cognitive decline leading to dementia and is associated with decreased brain NAD+ levels. This has triggered an intense interest in boosting nicotinamide adenine dinucleotide (NAD+) levels to prevent dementia. We tested if the administration of the precursor of NAD+, nicotinamide mononucleotide (NMN), can prevent diabetes-induced memory deficits. Diabetes was induced in Sprague-Dawley rats by the administration of streptozotocin (STZ). After 3 months of diabetes, hippocampal NAD+ levels were decreased (p = 0.011). In vivo localized high-resolution proton magnetic resonance spectroscopy (MRS) of the hippocampus showed an increase in the levels of glucose (p < 0.001), glutamate (p < 0.001), gamma aminobutyric acid (p = 0.018), myo-inositol (p = 0.018), and taurine (p < 0.001) and decreased levels of N-acetyl aspartate (p = 0.002) and glutathione (p < 0.001). There was a significant decrease in hippocampal CA1 neuronal volume (p < 0.001) and neuronal number (p < 0.001) in the Diabetic rats. Diabetic rats showed hippocampal related memory deficits. Intraperitoneal NMN (100 mg/kg) was given after induction and confirmation of diabetes and was provided on alternate days for 3 months. NMN increased brain NAD+ levels, normalized the levels of glutamate, taurine, N-acetyl aspartate (NAA), and glutathione. NMN-treatment prevented the loss of CA1 neurons and rescued the memory deficits despite having no significant effect on hyperglycemic or lipidemic control. In hippocampal protein extracts from Diabetic rats, SIRT1 and PGC-1α protein levels were decreased, and acetylation of proteins increased. NMN treatment prevented the diabetes-induced decrease in both SIRT1 and PGC-1α and promoted deacetylation of proteins. Our results indicate that NMN increased brain NAD+, activated the SIRT1 pathway, preserved mitochondrial oxidative phosphorylation (OXPHOS) function, prevented neuronal loss, and preserved cognition in Diabetic rats.
Collapse
Affiliation(s)
- Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Joungil Choi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA;
| | - Muhammed Ikbal Arvas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Mohammad Salimian
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Sujal Singh
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.X.); (R.P.G.)
| | - Rao P Gullapalli
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (S.X.); (R.P.G.)
| | - Tibor Kristian
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA;
- Department of Anesthesiology; University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James William Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (J.C.); (M.I.A.); (M.S.); (S.S.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA;
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence:
| |
Collapse
|
5
|
Manfredi G, Colombo E, Barsotti J, Benfenati F, Lanzani G. Photochemistry of Organic Retinal Prostheses. Annu Rev Phys Chem 2019; 70:99-121. [DOI: 10.1146/annurev-physchem-042018-052445] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Organic devices are attracting considerable attention as prostheses for the recovery of retinal light sensitivity lost to retinal degenerative disease. The biotic/abiotic interface created when light-sensitive polymers and living tissues are placed in contact allows excitation of a response in blind laboratory rats exposed to visual stimuli. Although polymer retinal prostheses have proved to be efficient, their working mechanism is far from being fully understood. In this review article, we discuss the results of the studies conducted on these kinds of polymer devices and compare them with the data found in the literature for inorganic retinal prostheses, where the working mechanisms are better comprehended. This comparison, which tries to set some reference values and figures of merit, is intended for use as a starting point to determine the direction for further investigation.
Collapse
Affiliation(s)
- Giovanni Manfredi
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, 20133 Milan, Italy;,
| | - Elisabetta Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genoa, Italy;,
| | - Jonathan Barsotti
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, 20133 Milan, Italy;,
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genoa, Italy;,
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
| | - Guglielmo Lanzani
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, 20133 Milan, Italy;,
- Department of Physics, Politecnico di Milano, 20133 Milan, Italy
| |
Collapse
|
6
|
de Carvalho MAJ, Chaves-Filho A, de Souza AG, de Carvalho Lima CN, de Lima KA, Rios Vasconcelos ER, Feitosa ML, Souza Oliveira JV, de Souza DAA, Macedo DS, de Souza FCF, de França Fonteles MM. Proconvulsant effects of sildenafil citrate on pilocarpine-induced seizures: Involvement of cholinergic, nitrergic and pro-oxidant mechanisms. Brain Res Bull 2019; 149:60-74. [PMID: 31004733 DOI: 10.1016/j.brainresbull.2019.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 03/26/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022]
Abstract
Sildenafil is a phosphodiesterase 5 inhibitor used for the treatment of erectile dysfunction and pulmonary hypertension. Proconvulsant effect is a serious adverse event associated with sildenafil use. Here, we investigated the possible proconvulsant effects of sildenafil in pilocarpine (PILO)-induced seizures model, which mimics some aspects of temporal lobe epilepsy. We also evaluated sildenafil's effects on hippocampal markers related to PILO-induced seizure, for instance, acetylcholinesterase (AChE) activity, oxidative stress and nitric oxide (NO) markers, namely nitrite, inducible NO synthase (iNOS) and neuronal NOS (nNOS). The influences of muscarinic receptors blockade on sildenafil proconvulsant effects and brain nitrite levels were also evaluated. Male mice were submitted to single or repeated (7 days) sildenafil administration (2.5, 5, 10 and 20 mg/kg). Thirty minutes later, PILO was injected and mice were further evaluated for 1 h for seizure activity. Sildenafil induced a dose- and time-progressive proconvulsant effect in PILO-induced seizures. Sildenafil also potentiated the inhibitory effect of PILO in AChE activity and induced a further increase in nitrite levels and pro-oxidative markers, mainly in the hippocampus. Repeated sildenafil treatment also increased the hippocampal expression of iNOS and nNOS isoforms, while the blockade of muscarinic receptors attenuated both sildenafil-induced proconvulsant effect and brain nitrite changes. Our data firstly demonstrated the proconvulsant effect of sildenafil in PILO-model of seizures. This effect seems to be related to an increased cholinergic-nitrergic tone and pro-oxidative brain changes. Also, our findings advert to caution in using sildenafil for patients suffering from neurological conditions that reduces seizure threshold, such as epilepsy.
Collapse
Affiliation(s)
- Michele Albuquerque Jales de Carvalho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Adriano Chaves-Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Alana Gomes de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Camila Nayane de Carvalho Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Klistenes Alves de Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Emiliano Ricardo Rios Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Mariana Lima Feitosa
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Victor Souza Oliveira
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Denia Alves Albuquerque de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Danielle S Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| | - Francisca Cléa Florenço de Souza
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Marta Maria de França Fonteles
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Department of Physiology and Pharmacology, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Pharmacy Department, Faculty of Dentistry, Nursing and Pharmacy, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
7
|
Spampinato SF, Copani A, Nicoletti F, Sortino MA, Caraci F. Metabotropic Glutamate Receptors in Glial Cells: A New Potential Target for Neuroprotection? Front Mol Neurosci 2018; 11:414. [PMID: 30483053 PMCID: PMC6243036 DOI: 10.3389/fnmol.2018.00414] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders are characterized by excitotoxicity and neuroinflammation that finally lead to slow neuronal degeneration and death. Although neurons are the principal target, glial cells are important players as they contribute by either exacerbating or dampening the events that lead to neuroinflammation and neuronal damage. A dysfunction of the glutamatergic system is a common event in the pathophysiology of these diseases. Metabotropic glutamate (mGlu) receptors belong to a large family of G protein-coupled receptors largely expressed in neurons as well as in glial cells. They often appear overexpressed in areas involved in neurodegeneration, where they can modulate glutamatergic transmission. Of note, mGlu receptor upregulation may involve microglia or, even more frequently, astrocytes, where their activation causes release of factors potentially able to influence neuronal death. The expression of mGlu receptors has been also reported on oligodendrocytes, a glial cell type specifically involved in the development of multiple sclerosis. Here we will provide a general overview on the possible involvement of mGlu receptors expressed on glial cells in the pathogenesis of different neurodegenerative disorders and the potential use of subtype-selective mGlu receptor ligands as candidate drugs for the treatment of neurodegenerative disorders. Negative allosteric modulators (NAM) of mGlu5 receptors might represent a relevant pharmacological tool to develop new neuroprotective strategies in these diseases. Recent evidence suggests that targeting astrocytes and microglia with positive allosteric modulators (PAM) of mGlu3 receptor or oligodendrocytes with mGlu4 PAMS might represent novel pharmacological approaches for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Agata Copani
- Department of Drug Sciences, University of Catania, Catania, Italy.,Institute of Biostructure and Bioimaging, National Research Council, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neuromed, Istituto di Ricovero e Cura a Carattere Scientifico, Pozzilli, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy.,Oasi Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico, Troina, Italy
| |
Collapse
|
8
|
Sita G, Hrelia P, Graziosi A, Ravegnini G, Morroni F. TRPM2 in the Brain: Role in Health and Disease. Cells 2018; 7:cells7070082. [PMID: 30037128 PMCID: PMC6070997 DOI: 10.3390/cells7070082] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 01/02/2023] Open
Abstract
Transient receptor potential (TRP) proteins have been implicated in several cell functions as non-selective cation channels, with about 30 different mammalian TRP channels having been recognized. Among them, TRP-melastatin 2 (TRPM2) is particularly involved in the response to oxidative stress and inflammation, while its activity depends on the presence of intracellular calcium (Ca2+). TRPM2 is involved in several physiological and pathological processes in the brain through the modulation of multiple signaling pathways. The aim of the present review is to provide a brief summary of the current insights of TRPM2 role in health and disease to focalize our attention on future potential neuroprotective strategies.
Collapse
Affiliation(s)
- Giulia Sita
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Agnese Graziosi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Fabiana Morroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| |
Collapse
|
9
|
Fernandes LF, Bruch GE, Massensini AR, Frézard F. Recent Advances in the Therapeutic and Diagnostic Use of Liposomes and Carbon Nanomaterials in Ischemic Stroke. Front Neurosci 2018; 12:453. [PMID: 30026685 PMCID: PMC6041432 DOI: 10.3389/fnins.2018.00453] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022] Open
Abstract
The complexity of the central nervous system (CNS), its limited self-repairing capacity and the ineffective delivery of most CNS drugs to the brain contribute to the irreversible and progressive nature of many neurological diseases and also the severity of the outcome. Therefore, neurological disorders belong to the group of pathologies with the greatest need of new technologies for diagnostics and therapeutics. In this scenario, nanotechnology has emerged with innovative and promising biomaterials and tools. This review focuses on ischemic stroke, being one of the major causes of death and serious long-term disabilities worldwide, and the recent advances in the study of liposomes and carbon nanomaterials for therapeutic and diagnostic purposes. Ischemic stroke occurs when blood flow to the brain is insufficient to meet metabolic demand, leading to a cascade of physiopathological events in the CNS including local blood brain barrier (BBB) disruption. However, to date, the only treatment approved by the FDA for this pathology is based on the potentially toxic tissue plasminogen activator. The techniques currently available for diagnosis of stroke also lack sensitivity. Liposomes and carbon nanomaterials were selected for comparison in this review, because of their very distinct characteristics and ranges of applications. Liposomes represent a biomimetic system, with composition, structural organization and properties very similar to biological membranes. On the other hand, carbon nanomaterials, which are not naturally encountered in the human body, exhibit new modes of interaction with biological molecules and systems, resulting in unique pharmacological properties. In the last years, several neuroprotective agents have been evaluated under the encapsulated form in liposomes, in experimental models of stroke. Effective drug delivery to the brain and neuroprotection were achieved using stealth liposomes bearing targeting ligands onto their surface for brain endothelial cells and ischemic tissues receptors. Carbon nanomaterials including nanotubes, fullerenes and graphene, started to be investigated and potential applications for therapy, biosensing and imaging have been identified based on their antioxidant action, their intrinsic photoluminescence, their ability to cross the BBB, transitorily decrease the BBB paracellular tightness, carry oligonucleotides and cells and induce cell differentiation. The potential future developments in the field are finally discussed.
Collapse
Affiliation(s)
| | | | - André R. Massensini
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Frédéric Frézard
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
10
|
Beytut E, Yilmaz S, Aksakal M, Polat S. The possible protective effects of vitamin E and selenium administration in oxidative stress caused by high doses of glucocorticoid administration in the brain of rats. J Trace Elem Med Biol 2018; 45:131-135. [PMID: 29173469 DOI: 10.1016/j.jtemb.2017.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/22/2017] [Accepted: 10/11/2017] [Indexed: 11/17/2022]
Abstract
Acute exposure to high doses of glucocorticoids (GCs) may potentially increase the basal levels of reactive oxygen species (ROS) by altering the defence capacity against oxidative damage. Also, antioxidants may affect the oxidative breakdown of tissues. Therefore, the aim of this work was to determine the effects of dietary intake vitamin E and selenium (Se) on lipid peroxidation (LPO) as thiobarbituric acid reactive substances (TBARS) and on the antioxidative defence mechanisms in the brain of rats treated with high doses of prednisolone. Two hundred and fifty adult male Wistar rats were randomly divided into five groups. The rats were fed a normal diet, but groups 3, 4, and 5 received a daily supplement in their drinking water of 20mg vitamin E, 0.3mg Se, and a combination of vitamin E and Se, respectively, for 30days. For 3days subsequently, the control (group 1) was treated with a placebo, and the remaining 4 groups were injected intramuscularly with 100mg/kg body weight (bw) prednisolone. After the last administration of prednisolone, 10 rats from each group were killed at 4, 8, 12, 24, and 48h and the activities of enzymes selenium-glutathione peroxidase (Se-GSH-Px) and catalase (CAT), and the levels of reduced glutathione (reduced GSH) and TBARS in their brains were measured. Se-GSH-Px and CAT enzyme activities, and reduced GSH levels in the prednisolone treatment group (group 2) began to decrease gradually at 4h (p<0.01, p<0.05, respectively), falling respectively to 60, 50, and 40% of the control levels by 24h (p<0.001, p<0.01), and recovering to the control levels at 48h. In contrast, prednisolone administration caused an increase in the brain TBARS, reaching up to six times the level of the control at 24h (p<0.001). However, supplementation with vitamin E and Se had a preventive effect on the elevation of the brain TBARS and improved the diminished activities of antioxidative enzymes and the levels of reduced GSH. Therefore, the present study attempts to determine the sequence of cellular membrane damage in the brain of the rats after high doses GC administration and the possible roles in vivo of vitamin E and Se, and their combination.
Collapse
Affiliation(s)
- Ebru Beytut
- Department of Medical Physiology, Faculty of Medicine, University of Erzincan, Erzincan, TURKEY
| | - Seval Yilmaz
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Firat, Elazıg, TURKEY
| | - Mesut Aksakal
- Department of Physiology, Faculty of Veterinary Medicine, University of Firat, Elazıg, TURKEY
| | - Seher Polat
- Department of Medical Genetics, Faculty of Medicine, University of Erzincan, Erzincan, TURKEY.
| |
Collapse
|
11
|
Abstract
Acute trauma can lead to life-long changes in susceptibility to psychiatric disease, such as post-traumatic stress disorder (PTSD). Rats given free access to a concentrated glucose solution for 24 h beginning immediately after trauma failed to show stress-related pathology in the learned helplessness model of PTSD and comorbid major depression. We assessed effective dosing and temporal constraints of the glucose intervention in three experiments. We exposed 120 male Sprague-Dawley rats to 100, 1 mA, 3-15 s, inescapable and unpredictable electric tail shocks (over a 110-min period) or simple restraint in the learned helplessness procedure. Rats in each stress condition had access to a 40% glucose solution or water. We measured fluid consumption under 18-h free access conditions, or limited access (1, 3, 6, 18 h) beginning immediately after trauma, or 3-h access with delayed availability of the glucose solution (0, 1, 3, 6 h). We hypothesized that longer and earlier access following acute stress would improve shuttle-escape performance. Rats exposed to traumatic shock and given 18-h access to glucose failed to show exaggerated fearfulness and showed normal reactivity to foot shock during testing as compared to their water-treated counterparts. At least 3 h of immediate post-stress access to glucose were necessary to see these improvements in test performance. Moreover, delaying access to glucose for more than 3 h post-trauma yielded no beneficial effects. These data clearly identify limits on the post-stress glucose intervention. In conclusion, glucose should be administered almost immediately and at the highest dose after trauma.
Collapse
Affiliation(s)
- M A Conoscenti
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
| | - E E Hart
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
| | - N J Smith
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
| | - T R Minor
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
- b UCLA Behavioral Testing Core , Brain Research Institute , Los Angeles , CA , USA
- c Department of Psychiatry and Biobehavioral Sciences , UCLA Integrative Center for Learning and Memory , Los Angeles , CA , USA
- d Stress and Motivated Behavior Institute , New Jersey Medical School , Newark , NJ , USA
| |
Collapse
|
12
|
Fang Y, Huang X, Wan Y, Tian H, Tian Y, Wang W, Zhu S, Xie M. Deficiency of TREK-1 potassium channel exacerbates secondary injury following spinal cord injury in mice. J Neurochem 2017; 141:236-246. [PMID: 28192611 DOI: 10.1111/jnc.13980] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/09/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Yongkang Fang
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Xiaojiang Huang
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Yue Wan
- Department of Neurology; The Third People's Hospital of Hubei Province; Wuhan China
| | - Hao Tian
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Yeye Tian
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Wei Wang
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
- Key Laboratory of Neurological Diseases of Chinese Ministry of Education; The School of Basic Medicine; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Suiqiang Zhu
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Minjie Xie
- Department of Neurology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
- Key Laboratory of Neurological Diseases of Chinese Ministry of Education; The School of Basic Medicine; Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| |
Collapse
|
13
|
Lee JH, Zhang J, Yu SP. Neuroprotective mechanisms and translational potential of therapeutic hypothermia in the treatment of ischemic stroke. Neural Regen Res 2017; 12:341-350. [PMID: 28469636 PMCID: PMC5399699 DOI: 10.4103/1673-5374.202915] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Stroke is a leading cause of disability and death, yet effective treatments for acute stroke has been very limited. Thus far, tissue plasminogen activator has been the only FDA-approved drug for thrombolytic treatment of ischemic stroke patients, yet its application is only applicable to less than 4–5% of stroke patients due to the narrow therapeutic window (< 4.5 hours after the onset of stroke) and the high risk of hemorrhagic transformation. Emerging evidence from basic and clinical studies has shown that therapeutic hypothermia, also known as targeted temperature management, can be a promising therapy for patients with different types of stroke. Moreover, the success in animal models using pharmacologically induced hypothermia (PIH) has gained increasing momentum for clinical translation of hypothermic therapy. This review provides an updated overview of the mechanisms and protective effects of therapeutic hypothermia, as well as the recent development and findings behind PIH treatment. It is expected that a safe and effective hypothermic therapy has a high translational potential for clinical treatment of patients with stroke and other CNS injuries.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| | - James Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA; Veteran's Affair Medical Center, Center for Visual and Neurocognitive Rehabilitation, Atlanta, GA, USA
| |
Collapse
|
14
|
Geevasinga N, Menon P, Özdinler PH, Kiernan MC, Vucic S. Pathophysiological and diagnostic implications of cortical dysfunction in ALS. Nat Rev Neurol 2016; 12:651-661. [DOI: 10.1038/nrneurol.2016.140] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
15
|
Tatter SB, Galpern WR, Isacson O. Neurotrophic Factor Protection against Excitotoxic Neuronal Death. Neuroscientist 2016. [DOI: 10.1177/107385849500100506] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neurotrophic factors are polypeptides capable of promoting neuronal survival in both the developing and the adult brain. In addition to the neurotrophins, NGF, brain-derived neurotropic factor, and NT-3 to -6, other neurotrophic factors include ciliary neurotrophic factor, fibroblast growth factors, insulin-like growth factors, members of the transforming growth factor superfamily, members of the epidermal growth factor family, and other cytokines such as leukemia inhibitory factor, oncostatin M, and interleukins-6 and -11. One condition under which these factors promote survival is the challenge of neurons with analogs of excitatory amino acid transmitters. Such analogs, including quinolinic acid, kainic acid, and ibotenic acid, are frequently employed as models of neurological diseases such as Huntington's disease, Parkinson's disease, Alzheimer's disease, epilepsy, cerebellar degenerations, and amyotrophic lateral sclerosis. Excitotoxicity also plays a role in neu ronal death caused by focal ischemia, hypoglycemia, or trauma. Although much has been learned about the mechanisms of both the action of neurotrophic factors and of cell death in response to excitotoxins, the mechanism of protection by these factors remains uncertain. This review explores the biochemical and phys iological changes mediated by neurotrophic factors that may underlie their ability to protect against excito toxic cell death. Second messenger pathways used degenerately by both excitotoxins and neurotrophic factors are discussed as a potential site of interaction mediating the protective effects of neurotrophic factors. Particular attention is also paid to the importance of the route of neurotrophic factor delivery in conferring neuroprotection in particular excitotoxic models. The Neuroscientist 1:286-297, 1995
Collapse
Affiliation(s)
- Stephen B. Tatter
- Departments of Neurosurgery and Neurology Massachusetts
General Hospital Boston, Massachusetts, Neuroregeneration Laboratory McLean Hospital Belmont,
Massachusetts
| | - Wendy R. Galpern
- Departments of Neurosurgery and Neurology Massachusetts
General Hospital Boston, Massachusetts, Neuroregeneration Laboratory McLean Hospital Belmont,
Massachusetts
| | - Ole Isacson
- Departments of Neurosurgery and Neurology Massachusetts
General Hospital Boston, Massachusetts, Neuroregeneration Laboratory McLean Hospital Belmont,
Massachusetts
| |
Collapse
|
16
|
|
17
|
Hypoxia-Induced Iron Accumulation in Oligodendrocytes Mediates Apoptosis by Eliciting Endoplasmic Reticulum Stress. Mol Neurobiol 2015; 53:4713-27. [PMID: 26319559 DOI: 10.1007/s12035-015-9389-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/11/2015] [Indexed: 10/23/2022]
Abstract
This study was aimed at evaluating the role of increased iron accumulation in oligodendrocytes and its role in their apoptosis in the periventricular white matter damage (PWMD) following a hypoxic injury to the neonatal brain. In response to hypoxia, in the PWM, there was increased expression of proteins involved in iron acquisition, such as iron regulatory proteins (IRP1, IRP2) and transferrin receptor in oligodendrocytes. Consistent with this, following a hypoxic exposure, there was increased accumulation of iron in primary cultured oligodendrocytes. The increased concentration of iron within hypoxic oligodendrocytes was found to elicit ryanodine receptor (RyR) expression, and the expression of endoplasmic reticulum (ER) stress markers such as binding-immunoglobulin protein (BiP) and inositol-requiring enzyme (IRE)-1α. Associated with ER stress, there was reduced adenosine triphosphate (ATP) levels within hypoxic oligodendrocytes. However, treatment with deferoxamine reduced the increased expression of RyR, BiP, and IRE-1α and increased ATP levels in hypoxic oligodendrocytes. Parallel to ER stress there was enhanced reactive oxygen species production within mitochondria of hypoxic oligodendrocytes, which was attenuated when these cells were treated with deferoxamine. At the ultrastructural level, hypoxic oligodendrocytes frequently showed dilated ER and disrupted mitochondria, which became less evident in those treated with deferoxamine. Associated with these subcellular changes, the apoptosis of hypoxic oligodendrocytes was evident with an increase in p53 and caspase-3 expression, which was attenuated when these cells were treated with deferoxamine. Thus, the present study emphasizes that the excess iron accumulated within oligodendrocytes in hypoxic PWM could result in their death by eliciting ER stress and mitochondrial disruption.
Collapse
|
18
|
Neurogenic neuroinflammation: inflammatory CNS reactions in response to neuronal activity. Nat Rev Neurosci 2013; 15:43-53. [PMID: 24281245 DOI: 10.1038/nrn3617] [Citation(s) in RCA: 388] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The CNS is endowed with an elaborated response repertoire termed 'neuroinflammation', which enables it to cope with pathogens, toxins, traumata and degeneration. On the basis of recent publications, we deduce that orchestrated actions of immune cells, vascular cells and neurons that constitute neuroinflammation are not only provoked by pathological conditions but can also be induced by increased neuronal activity. We suggest that the technical term 'neurogenic neuroinflammation' should be used for inflammatory reactions in the CNS in response to neuronal activity. We believe that neurogenic neuro-inflammation maintains homeostasis to enable the CNS to cope with enhanced metabolic demands and increases the computational power and plasticity of CNS neuronal networks. However, neurogenic neuroinflammation may also become maladaptive and aggravate the outcomes of pain, stress and epilepsy.
Collapse
|
19
|
Vucic S, Ziemann U, Eisen A, Hallett M, Kiernan MC. Transcranial magnetic stimulation and amyotrophic lateral sclerosis: pathophysiological insights. J Neurol Neurosurg Psychiatry 2013; 84:1161-70. [PMID: 23264687 PMCID: PMC3786661 DOI: 10.1136/jnnp-2012-304019] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder of the motor neurons in the motor cortex, brainstem and spinal cord. A combination of upper and lower motor neuron dysfunction comprises the clinical ALS phenotype. Although the ALS phenotype was first observed by Charcot over 100 years ago, the site of ALS onset and the pathophysiological mechanisms underlying the development of motor neuron degeneration remain to be elucidated. Transcranial magnetic stimulation (TMS) enables non-invasive assessment of the functional integrity of the motor cortex and its corticomotoneuronal projections. To date, TMS studies have established motor cortical and corticospinal dysfunction in ALS, with cortical hyperexcitability being an early feature in sporadic forms of ALS and preceding the clinical onset of familial ALS. Taken together, a central origin of ALS is supported by TMS studies, with an anterograde transsynaptic mechanism implicated in ALS pathogenesis. Of further relevance, TMS techniques reliably distinguish ALS from mimic disorders, despite a compatible peripheral disease burden, thereby suggesting a potential diagnostic utility of TMS in ALS. This review will focus on the mechanisms underlying the generation of TMS measures used in assessment of cortical excitability, the contribution of TMS in enhancing the understanding of ALS pathophysiology and the potential diagnostic utility of TMS techniques in ALS.
Collapse
Affiliation(s)
- Steve Vucic
- Sydney Medical School Westmead, University of Sydney, Sydney, New South Wales, Australia.
| | | | | | | | | |
Collapse
|
20
|
Chang HJ, Kim JM, Lee JC, Kim WK, Chun HS. Protective effect of β-caryophyllene, a natural bicyclic sesquiterpene, against cerebral ischemic injury. J Med Food 2013; 16:471-80. [PMID: 23734999 DOI: 10.1089/jmf.2012.2283] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
β-Caryophyllene (trans-4,11,11-trimethyl-8-methylenebicyclo[7,2,0]undec-4-ene), found in various plants, is a natural bicyclic sesquiterpene with a low toxicity. Here, we show that a single intraperitoneal injection of β-caryophyllene (10 mg/kg) significantly reduced the cortical infarct volume by 67% when given immediately before middle cerebral artery occlusion (MCAO). Neurological deficits caused by MCAO were also significantly decreased by β-caryophyllene. β-Caryophyllene treatment of cortical cells exposed to oxygen-glucose deprivation revealed a significant protection in a dose-dependent manner. However, β-caryophyllene neither suppressed N-methyl-D-aspartate excitotoxicity in cultured cortical cells nor markedly decreased the oxidative stress measured in the cellular or acellular systems. By contrast, treatments with β-caryophyllene dose-dependently inhibited mRNA expression of inducible nitric oxide synthetase, interleukin (IL)-1β, IL-6, and cyclooxygenase 2 in C6 microglial cells, and decreased the level of nitric oxide and prostaglandin E₂ at a 100 μM concentration. All of these findings suggest that β-caryophyllene has a potent neuroprotective activity, and its neuroprotection may be partly related to the modulation of inflammatory mediators.
Collapse
Affiliation(s)
- Hyun-Joo Chang
- Korea Food Research Institute, Sungnam, Kyonggi-do, Republic of Korea
| | | | | | | | | |
Collapse
|
21
|
Canzoniero LMT, Granzotto A, Turetsky DM, Choi DW, Dugan LL, Sensi SL. nNOS(+) striatal neurons, a subpopulation spared in Huntington's Disease, possess functional NMDA receptors but fail to generate mitochondrial ROS in response to an excitotoxic challenge. Front Physiol 2013; 4:112. [PMID: 23720635 PMCID: PMC3655281 DOI: 10.3389/fphys.2013.00112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 04/30/2013] [Indexed: 12/26/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative condition characterized by severe neuronal loss in the cortex and striatum that leads to motor and behavioral deficits. Excitotoxicity is thought to be involved in HD and several studies have indicated that NMDA receptor (NMDAR) overactivation can play a role in the selective neuronal loss found in HD. Interestingly, a small subset of striatal neurons (less than 1% of the overall population) is found to be spared in post-mortem HD brains. These neurons are medium-sized aspiny interneurons that highly express the neuronal isoform of nitric oxide synthase (nNOS). Intriguingly, neurons expressing large amounts of nNOS [hereafter indicated as nNOS(+) neurons] show reduced vulnerability to NMDAR-mediated excitotoxicity. Mechanisms underlying this reduced vulnerability are still largely unknown and may shed some light on pathogenic mechanisms involved in HD. One untested possibility is that nNOS(+) neurons possess fewer or less functioning NMDARs. Employing single cell calcium imaging we challenged this hypothesis and found that cultured striatal nNOS(+) neurons show NMDAR-evoked responses that are identical to the ones observed in the overall population of neurons that express lower levels of nNOS [nNOS(−) neurons]. NMDAR-dependent deregulation of intraneuronal Ca2+ is known to generate high levels of reactive oxygen species of mitochondrial origin (mt-ROS), a crucial step in the excitotoxic cascade. With confocal imaging and dihydrorhodamine (DHR; a ROS-sensitive probe) we compared mt-ROS levels generated by NMDAR activation in nNOS(+) and (−) cultured striatal neurons. DHR experiments revealed that nNOS(+) neurons failed to produce significant amounts of mt-ROS in response to NMDA exposure, thereby providing a potential mechanism for their reduced vulnerability to excitotoxicity and decreased vulnerability in HD.
Collapse
Affiliation(s)
- Lorella M T Canzoniero
- Department of Biological and Environmental Science, University of Sannio Benevento, Italy
| | | | | | | | | | | |
Collapse
|
22
|
New insights in mitochondrial calcium handling by sodium/calcium exchanger. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 961:203-9. [PMID: 23224881 DOI: 10.1007/978-1-4614-4756-6_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mitochondria are now recognized as one of the main intracellular calcium-storing organelles which play a key role in the intracellular calcium signalling. Indeed, besides performing oxidative phosphorylation, mitochondria are able to sense and shape calcium (Ca(2+)) transients, thus controlling cytosolic Ca(2+) signals and Ca(2+)-dependent protein activity. It has been well established for many years that mitochondria have a huge capacity to accumulate calcium. While the physiological significance of this pathway was hotly debated until relatively recently, it is now clear that the ability of mitochondria in calcium handling is a ubiquitous phenomenon described in every cell system in which the issue has been addressed.In this chapter, we will review the molecular mechanisms involved in the regulation of mitochondrial calcium cycling in physiological conditions with particular regard to the role played by the mitochondrial Na(+)/Ca(2+) exchanger.
Collapse
|
23
|
Vucic S, Kiernan MC. Utility of transcranial magnetic stimulation in delineating amyotrophic lateral sclerosis pathophysiology. HANDBOOK OF CLINICAL NEUROLOGY 2013; 116:561-575. [PMID: 24112924 DOI: 10.1016/b978-0-444-53497-2.00045-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disorder of the motor neurons in the motor cortex, brainstem, and spinal cord. The clinical phenotype of ALS is underscored by a combination of upper and lower motor neuron dysfunction. Although this phenotype was observed over 100 years ago, the site of ALS onset and the pathophysiological mechanisms underlying the development of motor neuron degeneration remain to be elucidated. Transcranial magnetic stimulation (TMS) enables noninvasive assessment of the functional integrity of the motor cortex and its corticomotoneuronal projections. To date, TMS studies have established cortical dysfunction in ALS, with cortical hyperexcitability being an early feature in sporadic forms of ALS and preceding the clinical onset of familial ALS. Taken together, a central origin of ALS is supported by TMS studies, with an anterograde dying-forward mechanism implicated in ALS pathogenesis. Of further relevance, TMS techniques reliably distinguish ALS from mimic disorders, despite a compatible peripheral disease burden, thereby suggesting a potential diagnostic utility of TMS in ALS. This chapter reviews the mechanisms underlying the generation of TMS parameters utilized in assessment of cortical excitability, the contribution of TMS in enhancing the understanding of ALS pathophysiology, and the potential diagnostic utility of TMS techniques in ALS.
Collapse
Affiliation(s)
- Steve Vucic
- Sydney Medical School Westmead, University of Sydney, Sydney, Australia; Neuroscience Research Australia, Sydney, Australia
| | | |
Collapse
|
24
|
Falcão HDS, Maia GLDA, Bonamin F, Kushima H, Moraes TM, Hiruma Lima CA, Takayama C, Ferreira AL, Souza Brito ARM, Agra MDF, Barbosa Filho JM, Batista LM. Gastroprotective mechanisms of the chloroform and ethyl acetate phases of Praxelis clematidea (Griseb.) R.M.King & H.Robinson (Asteraceae). J Nat Med 2012; 67:480-91. [DOI: 10.1007/s11418-012-0705-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/21/2012] [Indexed: 01/22/2023]
|
25
|
Borgens RB, Liu-Snyder P. Understanding secondary injury. QUARTERLY REVIEW OF BIOLOGY 2012; 87:89-127. [PMID: 22696939 DOI: 10.1086/665457] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Secondary injury is a term applied to the destructive and self-propagating biological changes in cells and tissues that lead to their dysfunction or death over hours to weeks after the initial insult (the "primary injury"). In most contexts, the initial injury is usually mechanical. The more destructive phase of secondary injury is, however, more responsible for cell death and functional deficits. This subject is described and reviewed differently in the literature. To biomedical researchers, systemic and tissue-level changes such as hemorrhage, edema, and ischemia usually define this subject. To cell and molecular biologists, "secondary injury" refers to a series of predominately molecular events and an increasingly restricted set of aberrant biochemical pathways and products. These biochemical and ionic changes are seen to lead to death of the initially compromised cells and "healthy" cells nearby through necrosis or apoptosis. This latter process is called "bystander damage." These viewpoints have largely dominated the recent literature, especially in studies of the central nervous system (CNS), often without attempts to place the molecular events in the context of progressive systemic and tissue-level changes. Here we provide a more comprehensive and inclusive discussion of this topic.
Collapse
Affiliation(s)
- Richard Ben Borgens
- Center for Paralysis Research, School of Veterinary Medicine, Department of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA.
| | | |
Collapse
|
26
|
Im DS, Jeon JW, Lee JS, Won SJ, Cho SI, Lee YB, Gwag BJ. Role of the NMDA receptor and iron on free radical production and brain damage following transient middle cerebral artery occlusion. Brain Res 2012; 1455:114-23. [PMID: 22483792 DOI: 10.1016/j.brainres.2012.03.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/08/2012] [Accepted: 03/09/2012] [Indexed: 11/28/2022]
Abstract
Excess activation of ionotropic glutamate receptors and iron is believed to contribute to free radical production and neuronal death following hypoxic ischemia. We examined the possibility that both NMDA receptor activation and iron overload determine spatial and temporal patterns of free radical production after transient middle cerebral artery occlusion (tMCAO) in male Sprague-Dawley rats. Mitochondrial free radical (MFR) levels were maximally increased in neurons in the core at 1 h and 24 h after tMCAO. Early MFR production was blocked by administration of MK-801, an NMDA receptor antagonist, but not deferoxamine, an iron chelator. Neither MK-801 nor deferoxamine attenuated late MFR production in the core. Increased MFRs were observed in penumbral neurons within 6 h and gradually increased over 24 h after tMCAO. Slowly-evolving MFRs in the core and penumbra were accompanied by iron overload. Deferoxamine blocked iron overload but reduced MFR production only in the penumbra. Combined MK-801/deferoxamine reduced late MFR production in both core and penumbra in an additive manner. Combination therapy significantly ameliorated infarction compared with monotherapy. These findings suggest that the NMDA receptor activation and iron overload mediate late MFR production and infarction after tMCAO.
Collapse
Affiliation(s)
- Doo Soon Im
- GNT Pharma Research Institute, Gomae-Dong 381-1, Giheung-Gu, Yongin-Si, 446-901, South Korea
| | | | | | | | | | | | | |
Collapse
|
27
|
Chung JY, Yi JW, Kim SM, Lim YJ, Chung JH, Jo DJ. Changes in gene expression in the rat hippocampus after focal cerebral ischemia. J Korean Neurosurg Soc 2011; 50:173-8. [PMID: 22102944 DOI: 10.3340/jkns.2011.50.3.173] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 07/19/2011] [Accepted: 09/14/2011] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE The rat middle cerebral artery thread-occlusion model has been widely used to investigate the pathophysiological mechanisms of stroke and to develop therapeutic treatment. This study was conducted to analyze energy metabolism, apoptotic signal pathways, and genetic changes in the hippocampus of the ischemic rat brain. METHODS Focal transient cerebral ischemia was induced by obstructing the middle cerebral artery for two hours. After 24 hours, the induction of ischemia was confirmed by the measurement of infarct size using 2,3,5-triphenyltetrazolium chloride staining. A cDNA microarray assay was performed after isolating the hippocampus, and was used to examine changes in genetic expression patterns. RESULTS According to the cDNA microarray analysis, a total of 1,882 and 2,237 genes showed more than a 2-fold increase and more than a 2-fold decrease, respectively. When the genes were classified according to signal pathways, genes related with oxidative phosphorylation were found most frequently. There are several apoptotic genes that are known to be expressed during ischemic brain damage, including Akt2 and Tnfrsf1a. In this study, the expression of these genes was observed to increase by more than 2-fold. As energy metabolism related genes grew, ischemic brain damage was affected, and the expression of important genes related to apoptosis was increased/decreased. CONCLUSION Our analysis revealed a significant change in the expression of energy metabolism related genes (Atp6v0d1, Atp5g2, etc.) in the hippocampus of the ischemic rat brain. Based on this data, we feel these genes have the potential to be target genes used for the development of therapeutic agents for ischemic stroke.
Collapse
Affiliation(s)
- Jun Young Chung
- Department of Anesthesiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
28
|
Park JW, Kwon HJ, Chung WS, Kim CY, Seong GJ. Short-term effects of Ginkgo biloba extract on peripapillary retinal blood flow in normal tension glaucoma. KOREAN JOURNAL OF OPHTHALMOLOGY 2011; 25:323-8. [PMID: 21976939 PMCID: PMC3178766 DOI: 10.3341/kjo.2011.25.5.323] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 10/06/2010] [Indexed: 12/03/2022] Open
Abstract
Purpose Based on the vascular theory of glaucoma pathogenesis, we wanted to evaluate the effect of Ginkgo biloba extract (GBE) on peripapillary blood flow in patients with normal tension glaucoma (NTG). Methods Thirty patients with NTG were randomly placed in the GBE-treated or control groups. The GBE-treated group received 80 mg GBE orally, twice a day for four weeks, and the control group received a placebo twice a day for four weeks. Complete ocular examinations including visual field, Heidelberg retina flowmeter, and systemic examinations were performed on the first study day and on the day treatment was completed. Results After GBE treatment, the mean blood flow, volume, and velocity increased at almost all points, and there was a statistically significant increase in blood flow at almost all points, in comparison to the placebo. Blood volume significantly increased only in the superior nasal and superior temporal neuroretinal rim areas. GBE also significantly increased blood velocity in areas of the inferior temporal neuroretinal rim and superior temporal peripapillary area. Conclusions GBE administration appears to have desirable effect on ocular blood flow in NTG patients.
Collapse
Affiliation(s)
- Jong Woon Park
- Department of Ophthalmology, NHIC Ilsan Hospital, Goyang, Korea.
| | | | | | | | | |
Collapse
|
29
|
Effect of Vitamin E on Blood-Brain Barrier Permeability in Aged Rats with PTZ-Induced Convulsions. NEUROPHYSIOLOGY+ 2011. [DOI: 10.1007/s11062-011-9168-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
30
|
Turunc E, Kanit L, Yalcin A. Effect of γ-glutamylcysteine ethylester on the levels of c-fos mRNA expression, glutathione and reactive oxygen species formation in kainic acid excitotoxicity. J Pharm Pharmacol 2010; 62:1010-7. [DOI: 10.1111/j.2042-7158.2010.01122.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Objectives
The aim of this study was to investigate the effect of γ-glutamylcysteine ethylester (GCEE), a precursor of glutathione biosynthesis, on the levels of glutathione, formation of reactive oxygen species and c-fos mRNA expression in rat hippocampus and cortex in kainic acid-induced excitotoxicity.
Methods
Sprague–Dawley rats were used and divided into four groups: control, kainic acid (10 mg/kg), GCEE (10 mg/kg) and kainic acid (10 mg/kg) + GCEE (10 mg/kg). Kainic acid and GCEE were administered to the rats intraperitoneally. The levels of glutathione and the expressions of c-fos mRNA in hippocampus and cortex tissues were determined using spectrophotometric and reverse transcription followed real-time PCR methods, respectively. Formation of reactive oxygen species was determined using dichlorofluorescin fluorescence in brain synaptosomes treated with kainic acid or GCEE in vitro.
Key findings
Kainic acid treatment significiantly upregulated the expression of c-fos mRNA in the hippocampus and cortex when compared to the control group. GCEE treatment significantly decreased the levels of c-fos mRNA in the cortex when compared to the kainic acid-treated group. GCEE treatment against kainic acid significantly increased the levels of glutathione in the cortex and hippocampus, and decreased the levels of formation of reactive oxygen species when compared to kainic acid-treated synaptosomes.
Conclusions
The increased levels of glutathione and the reduced levels of reactive oxygen species formation lead us to conclude that GCEE may be beneficial as a potential antioxidant against neurodegenerative processes where excitotoxicity is involved.
Collapse
Affiliation(s)
- Ezgi Turunc
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Lutfiye Kanit
- Department of Physiology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ayfer Yalcin
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
31
|
Kusama-Eguchi K, Yamazaki Y, Ueda T, Suda A, Hirayama Y, Ikegami F, Watanabe K, May M, Lambein F, Kusama T. Hind-limb paraparesis in a rat model for neurolathyrism associated with apoptosis and an impaired vascular endothelial growth factor system in the spinal cord. J Comp Neurol 2010; 518:928-42. [DOI: 10.1002/cne.22257] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Filipović D, Mandić LM, Kanazir D, Pajović SB. Acute and/or chronic stress models modulate CuZnSOD and MnSOD protein expression in rat liver. Mol Cell Biochem 2009; 338:167-74. [DOI: 10.1007/s11010-009-0350-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 12/03/2009] [Indexed: 01/11/2023]
|
33
|
Sun P, Gu J, Maze M, Ma D. Is xenon a future neuroprotectant? FUTURE NEUROLOGY 2009. [DOI: 10.2217/fnl.09.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Acute neuronal injury has devastating consequences with increased risks of morbidity and mortality. Among its survivors, neurological deficit is associated with loss of function, independence and quality of life. Currently, there is a distinctive lack of effective clinical strategies to obviate this problem. Xenon, a noble gas with anesthetic properties, exhibits neuroprotective effects. It is efficacious and nontoxic and has been used safely in clinical settings involving both anesthetic and imaging applications in patients of all ages. Xenon blocks the NMDA subtype of the glutamate receptor, a pivotal step in the pathway towards neuronal death. The preclinical data obtained from animal models of stroke, neonatal asphyxia and global ischemia induced by cardiac arrest, as well as recent data of traumatic brain injury, revealed that xenon is a potentially ideal candidate as a neuroprotectant. In addition, recent studies demonstrated that xenon can uniquely prevent anesthetic-induced neurodegeneration in the developing brain. Thus, clinical studies are urgently required to investigate the neuroprotective effects of xenon in the clinical setting of brain damage.
Collapse
Affiliation(s)
- Pamela Sun
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Jianteng Gu
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, Chelsea and Westminster Hospital, London, UK and, Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Mervyn Maze
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Daqing Ma
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London SW10 9NH, UK
| |
Collapse
|
34
|
Dentate granule cells form hilar basal dendrites in a rat model of hypoxia-ischemia. Brain Res 2009; 1285:182-7. [PMID: 19539612 DOI: 10.1016/j.brainres.2009.06.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 06/08/2009] [Accepted: 06/11/2009] [Indexed: 01/04/2023]
Abstract
Hilar basal dendrites form on dentate granule cells following seizures. To determine whether other brain insults cause the formation of hilar basal dendrites, a model of global cerebral hypoxia/ischemia was used. Rats underwent a transient induction of ischemia by occlusion of both common carotid arteries followed by reperfusion. Hippocampal slices were prepared from these animals 1 month after the ischemic insult, and granule cells were labeled with a retrograde tracing technique after biocytin injections into stratum lucidum of CA3b. Ischemic rats had numerous biocytin-labeled granule cells with hilar basal dendrites located at the hilar border of the granule cell layer. Quantitative analysis of ischemic rats compared to controls showed a significant increase in the percentage of biocytin-labeled granule cells with hilar basal dendrites. These data demonstrate that other brain insults in addition to epilepsy may result in the formation of hilar basal dendrites on granule cells.
Collapse
|
35
|
Brouns R, De Deyn PP. The complexity of neurobiological processes in acute ischemic stroke. Clin Neurol Neurosurg 2009; 111:483-95. [PMID: 19446389 DOI: 10.1016/j.clineuro.2009.04.001] [Citation(s) in RCA: 372] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 04/04/2009] [Accepted: 04/06/2009] [Indexed: 01/24/2023]
Abstract
There is an urgent need for improved diagnostics and therapeutics for acute ischemic stroke. This is the focus of numerous research projects involving in vitro studies, animal models and clinical trials, all of which are based on current knowledge of disease mechanisms underlying acute focal cerebral ischemia. Insight in the chain of events occurring during acute ischemic injury is essential for understanding current and future diagnostic and therapeutic approaches. In this review, we summarize the actual knowledge on the pathophysiology of acute ischemic stroke. We focus on the ischemic cascade, which is a complex series of neurochemical processes that are unleashed by transient or permanent focal cerebral ischemia and involves cellular bioenergetic failure, excitotoxicity, oxidative stress, blood-brain barrier dysfunction, microvascular injury, hemostatic activation, post-ischemic inflammation and finally cell death of neurons, glial and endothelial cells.
Collapse
Affiliation(s)
- R Brouns
- Department of Neurology and Memory Clinic, Middelheim General Hospital, Antwerp, Belgium
| | | |
Collapse
|
36
|
|
37
|
Xavier SM, Barbosa CO, Barros DO, Silva RF, Oliveira AA, Freitas RM. Vitamin C antioxidant effects in hippocampus of adult Wistar rats after seizures and status epilepticus induced by pilocarpine. Neurosci Lett 2007; 420:76-9. [PMID: 17512118 DOI: 10.1016/j.neulet.2007.04.056] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 04/20/2007] [Accepted: 04/23/2007] [Indexed: 11/15/2022]
Abstract
Vitamin C (VIT C) is an exogenous antioxidant able to alter the brain oxidative stress. Antioxidant properties have been showed in seizures and status epilepticus (SE) induced by pilocarpine in adult rats. This present study was aimed at was investigating the VIT C effects on latency to first seizure, in percentage of seizures, mortality rate, as well as hippocampal lipid peroxidation levels and catalase activity after seizures and SE. The VIT C effects were investigated after the pretreatment with dose 250 mg/kg, i.p., 30 min before pilocarpine administration (400mg/kg, s.c., pilocarpine group (P400)). The VIT C increase the latency to first seizure and decrease the mortality rate and lipid peroxidation levels. In P400+VIT C and VIT C groups were observed an increase in hippocampal catalase activity. Our results suggests that the vitamin C can exert antioxidant and anticonvulsive effects in adult rats, suggesting that this vitamin can be able by reduction of lipid peroxidation content and increased of catalase enzymatic activity which cerebral compensatory mechanisms in free radical formation during SE.
Collapse
Affiliation(s)
- S M Xavier
- Curso de Farmácia da Faculdade Católica Rainha do Sertão, 660, Centro, Quixadá, 63900-000, Brazil
| | | | | | | | | | | |
Collapse
|
38
|
Kim JM, Lee JC, Chang N, Chun HS, Kim WK. S-Allyl-L-cysteine attenuates cerebral ischemic injury by scavenging peroxynitrite and inhibiting the activity of extracellular signal-regulated kinase. Free Radic Res 2007; 40:827-35. [PMID: 17015261 DOI: 10.1080/10715760600719540] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
S-Allyl-L-cysteine (SAC) has been shown to reduce ischemic injury due to its antioxidant activity. However, the antioxidant property of SAC has been controversial. The present study investigated the neuroprotective mechanism of SAC in cerebral ischemic insults. SAC decreased the size of infarction after transient or global ischemic insults. While it did not alter the N-methyl-D-aspartate excitotoxicity, SAC significantly scavenged the endogenously or exogenously produced ONOO- and reduced ONOO- cytotoxicity. In contrast, SAC has much lower scavenging activity against H2O2, O2*(-) or NO. Further, SAC inhibited the activity of extracellular signal-regulated kinase (ERK) increased in cultured neurons exposed to oxygen-glucose deprivation or in rat brain tissue after transient middle cerebral artery occlusion. The neuroprotective effect of SAC was mimicked by the ERK inhibitor U0125. The present results indicate that SAC exert its neuroprotective effect by scavenging ONOO- and inhibiting the ERK signaling pathway activated during initial hypoxic/ischemic insults.
Collapse
Affiliation(s)
- Ji-Myung Kim
- Food Function Research Division, Korea Food Research Institute, Sungnam, South Korea
| | | | | | | | | |
Collapse
|
39
|
Oliveira AA, Almeida JPC, Freitas RM, Nascimento VS, Aguiar LMV, Júnior HVN, Fonseca FN, Viana GSB, Sousa FCF, Fonteles MMF. Effects of levetiracetam in lipid peroxidation level, nitrite-nitrate formation and antioxidant enzymatic activity in mice brain after pilocarpine-induced seizures. Cell Mol Neurobiol 2007; 27:395-406. [PMID: 17205390 DOI: 10.1007/s10571-006-9132-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2006] [Accepted: 11/08/2006] [Indexed: 10/23/2022]
Abstract
: Oxidative stress has been implicated in a large number of human degenerative diseases, including epilepsy. Levetiracetam (LEV) is a new antiepileptic agent with broad-spectrum effects on seizures and animal models of epilepsy. Recently, it was demonstrated that the mechanism of LEV differs from that of conventional antiepileptic drugs. Objectifying to investigate if LEV mechanism of action involves antioxidant properties, lipid peroxidation levels, nitrite-nitrate formation, catalase activity, and glutathione (GSH) content were measured in adult mice brain. The neurochemical analyses were carried out in hippocampus of animals pretreated with LEV (200 mg/kg, i.p.) 60 min before pilocarpine-induced seizures (400 mg/kg, s.c.). The administration of alone pilocarpine, 400 mg/kg, s.c. (P400) produced a significant increase of lipid peroxidation level in hippocampus. LEV pretreatment was able to counteract this increase, preserving the lipid peroxidation level in normal value. P400 administration also produced increase in the nitrite-nitrate formation and catalase activity in hippocampus, beyond a decrease in GSH levels. LEV administration before P400 prevented the P400-induced alteration in nitrite-nitrate levels and preserved normal values of catalase activity in hippocampus. Moreover, LEV administration prevented the P400-induced loss of GSH in this cerebral area. The present data suggest that the protective effects of LEV against pilocarpine-induced seizures can be mediated, at least in part, by reduction of lipid peroxidation and hippocampal oxidative stress.
Collapse
Affiliation(s)
- A A Oliveira
- Laboratory of Neuropharmacology, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Rua Cel. Nunes de Melo 1127, Fortaleza 60431-970, CE, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The implantation of exogenous cells or tissues has been a popular and successful strategy to overcome physical discontinuity and support axon growth in experimental models of spinal cord injury (SCI). Cellular therapies exhibit a multifarious potential for SCI restoration, providing not only a supportive substrate upon which axons can traverse the injury site, but also reducing progressive tissue damage and scarring, facilitating remyelination repair, and acting as a source for replacing and re-establishing lost neural tissue and its circuitry. The past two decades of research into cell therapies for SCI repair have seen the progressive evolution from whole tissue strategies, such as peripheral nerve grafts, to the use of specific, purified cell types from a diverse range of sources and, recently, to the employment of stem or neural precursor cell populations that have the potential to form a full complement of neural cell types. Although the progression of cell therapies from laboratory to clinical implementation has been slow, human SCI safety and efficacy trials involving several cell types within the US appear to be close at hand.
Collapse
Affiliation(s)
- Damien D Pearse
- University of Miami Miller School of Medicine, The Miami Project to Cure Paralysis, Department of Neurological Surgery, Lois Pope Life Center, 1095 NW 14th Terrace (R-48), Miami, FL 33136, USA.
| | | |
Collapse
|
41
|
Abrahám IM, Meerlo P, Luiten PGM. Concentration dependent actions of glucocorticoids on neuronal viability and survival. Dose Response 2006; 4:38-54. [PMID: 18648635 DOI: 10.2203/dose-response.004.01.004.abraham] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A growing body of evidence based on experimental data demonstrates that glucocorticoids (GCs) can play a potent role in the survival and death of neurons. However, these observations reflect paradoxical features of GCs, since these adrenal stress hormones are heavily involved in both neurodegenerative and neuroprotective processes. The actual level of GCs appears to have an essential impact in this bimodal action. In the present short review we aim to show the importance of concentration dependent action of GCs on neuronal cell viability and cell survival in the brain. Additionally, we will summarize the possible GC-induced cellular mechanisms at different GC concentrations providing a background for their effect on the fate of nerve cells in conditions that are a challenge to their survival.
Collapse
Affiliation(s)
- István M Abrahám
- Neurobiology Research Group, Hungarian Academy of Sciences at Eötvös Loránd University, Budapest, Hungary
| | | | | |
Collapse
|
42
|
Gupta R, Jovin TG, Krieger DW. Therapeutic hypothermia for stroke: do new outfits change an old friend? Expert Rev Neurother 2006; 5:235-46. [PMID: 15853493 DOI: 10.1586/14737175.5.2.235] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Clinically significant neuroprotection for the brain continues to be an elusive quest. All attempts at developing effective pharmacologic agents have failed in clinical trials. Hypothermia has been thought to confer protection after brain injury for many years, but has recently regained interest as a neuroprotectant for focal ischemic stroke in the basic science and clinical literature. The failure to develop safe and efficacious pharmacologic agents along with promising clinical data on the efficacy of hypothermia for cardiac arrest patients have raised a great interest in hypothermia as a neuroprotectant for ischemic stroke. As a clinically meaningful neuroprotectant for stroke, hypothermia confers several theoretical advantages over pharmacologic agents. A major problem with neuroprotectant therapy is instituting therapy within a narrow time window. This obstacle may be easier for hypothermia to overcome as emergency medical service personnel can theoretically initiate it in the field. Additionally, pharmacologic agents are usually restricted to one aspect of the pathophysiologic cascade triggered by focal ischemia, whereas hypothermia acts on several of these pathways simultaneously. The recent advances and future directions in the utilization of hypothermia as a potential therapy for focal ischemic stroke are reviewed.
Collapse
Affiliation(s)
- Rishi Gupta
- Department of Neurology, Stroke Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | | | | |
Collapse
|
43
|
Lim JH, Lee JC, Lee YH, Choi IY, Oh YK, Kim HS, Park JS, Kim WK. Simvastatin prevents oxygen and glucose deprivation/reoxygenation-induced death of cortical neurons by reducing the production and toxicity of 4-hydroxy-2E-nonenal. J Neurochem 2006; 97:140-50. [PMID: 16515553 DOI: 10.1111/j.1471-4159.2006.03715.x] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lipid membrane peroxidation is highly associated with neuronal death in various neurodegenerative diseases including cerebral stroke. Here, we report that simvastatin decreases oxygen and glucose deprivation (OGD)/reoxygenation-evoked neuronal death by inhibiting the production and cytoxicity of 4-hydroxy-2E-nonenal (HNE), the final product of lipid peroxidation. Simvastatin markedly decreased the OGD/reoxygenation-evoked death of cortical neurons. OGD/reoxygenation increased the intracellular HNE level mostly in neuronal cells, not glial cells. Simvastatin decreased the intracellular level of HNE in neuronal cells exposed to OGD/reoxygenation. We further found that HNE induced the cytotoxicity in neuronal cells and synergistically increased the N-methyl-D-aspartate (NMDA) receptor-mediated excitotoxicity. Simvastatin largely blocked the NMDA neurotoxicity potentiated by HNE. However, simvastatin did not alter the NMDA-evoked calcium influx in the absence or presence of HNE. HNE inhibited the activity of nuclear factor-kappa B (NF-kappaB), and the cytotoxicity of HNE was in good correlation with inactivation of NF-kappaB. Simvastatin reversed the inhibition of NF-kappaB activity induced by OGD/reoxygenation or HNE. The neuroprotection by simvastatin was significantly attenuated by various NF-kappaB inhibitors, implying that simvastatin inhibits the cytotoxicity of HNE at least in part by maintaining the activity of NF-kappaB. Further understanding of the neuroprotective mechanism of simvastatin may provide a therapeutic strategy for oxidative stress-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Ji Hyae Lim
- Department of Neuroscience, College of Medicine, Laboratory of neurodegenerative diseases, Ewha Medical Center, Ewha Women's University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Oxidative stress and DNA damage–DNA repair system in vascular smooth muscle cells in artery and vein grafts. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.jccr.2005.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
45
|
Skrzydlewska E, Augustyniak A, Michalak K, Farbiszewski R. Green tea supplementation in rats of different ages mitigates ethanol-induced changes in brain antioxidant abilities. Alcohol 2005; 37:89-98. [PMID: 16584972 DOI: 10.1016/j.alcohol.2005.12.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 12/20/2005] [Accepted: 12/21/2005] [Indexed: 12/11/2022]
Abstract
Oxidative stress induced by chronic ethanol consumption, particularly in aging subjects, has been implicated in the pathophysiology of many neurodegenerative diseases. Antioxidants with polyphenol structures, such as those contained in green tea, given alone for 5 weeks in liquid Lieber de Carli diet followed by administration with ethanol for 4 weeks with ethanol have been investigated as potential therapeutic antioxidant agents in the brain in rats of three ages (2, 12, and 24 months). Ethanol consumption caused age-dependent decreases in brain superoxide dismutase, glutathione peroxidase, glutathione reductase, and catalase activities. In addition, ethanol consumption caused age-dependent decreases in the levels of GSH, selenium, vitamins, E, A and C, and beta-carotene and increases in the levels of oxidized glutathione (GSSG). Changes in the brain's antioxidative ability were accompanied by enhanced oxidative modification of lipids (increases in lipid hydroperoxides, malondialdehyde, and 4-hydroxynonenal levels) and proteins (increases in carbonyl groups and bistyrosine). Reduced risk of oxidative stress and protection of the central nervous system, particularly in young and adult rats, after green tea supplementation were observed. Green tea partially prevented changes in antioxidant enzymatic as well as nonenzymatic parameters induced by ethanol and enhanced by aging. Administration of green tea significantly protects lipids and proteins against oxidative modifications in the brain tissue of young and adult rats. The beneficial effect of green tea can result from the inhibition of free radical chain reactions generated during ethanol-induced oxidative stress and/or from green tea-induced increases in antioxidative abilities made possible by increases in the activity/concentration of endogenous antioxidants.
Collapse
Affiliation(s)
- Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, 15-089 Białystok, Poland.
| | | | | | | |
Collapse
|
46
|
Rothstein RP, Levison SW. Gray matter oligodendrocyte progenitors and neurons die caspase-3 mediated deaths subsequent to mild perinatal hypoxic/ischemic insults. Dev Neurosci 2005; 27:149-59. [PMID: 16046849 PMCID: PMC1351040 DOI: 10.1159/000085987] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2004] [Accepted: 03/09/2005] [Indexed: 11/19/2022] Open
Abstract
With significant improvements in neonatal care, fewer infants sustain severe injury as a consequence of hypoxia/ischemia (H/I). However, the majority of experimental studies have inflicted moderate to severe injuries, or they have assessed damage to the caudal forebrain; therefore, to better understand how a mild H/I episode affects the structures and cells of the rostral forebrain, we assessed the relative vulnerabilities of cells in the neocortex, striatum, corpus callosum, choroid plexus and subventricular zone (SVZ). To inflict mild H/I injury, the right common carotid artery was ligated followed by 1 h of hypoxia (8% O(2)) at 37 degrees C. Regional vulnerabilities were assessed using TUNEL, active caspase-3 and hematoxylin and eosin staining at 24 and 48 h of recovery. Scattered columns of cell death were observed in the neocortex with deep-layer neurons more vulnerable than more superficial neurons. The majority of these dying neurons appeared to be dying apoptotic rather than necrotic deaths. In addition, approximately 1/3 of the apoptotic cells in the neocortex were O4+ oligodendrocyte progenitors. We also observed a decrease in NG2 staining within the affected regions of the forebrain. By contrast, active caspase-3+/S-100beta+ astrocytes were not observed. Neurons and O4+ oligodendrocyte progenitors also died apoptotic deaths within the striatum. The lining cells of the choroid plexus also sustained damage. Elevated numbers of apoptotic cells were observed in the most lateral region of the SVZ and some of these dying cells were O4+. The most novel finding of this study, that oligodendrocyte progenitors in the gray matter are damaged and eliminated as a consequence of perinatal H/I, provides new insights into the histopathology and neurological deficits observed in infants who sustain mild H/I brain injuries.
Collapse
Affiliation(s)
- Raymond P Rothstein
- Department of Neurology and Neuroscience, Laboratory for Regenerative Neurobiology, UMDNJ-New Jersey Medical School, Newark, NJ, USA
| | | |
Collapse
|
47
|
Nascimento VS, D'alva MS, Oliveira AA, Freitas RM, Vasconcelos SMM, Sousa FCF, Fonteles MMF. Antioxidant effect of nimodipine in young rats after pilocarpine-induced seizures. Pharmacol Biochem Behav 2005; 82:11-6. [PMID: 16115668 DOI: 10.1016/j.pbb.2005.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2005] [Revised: 07/05/2005] [Accepted: 07/07/2005] [Indexed: 10/25/2022]
Abstract
Nimodipine (ND) is a centrally active calcium antagonist that blocks the voltage-dependent L-type channels. Its antiepileptic properties have been proved in various animal models, including pilocarpine-induced seizures in adult rats. In order to investigate protective effects of the ND (10 (ND10) and 30 mg/kg (ND30), i.p.), young male rats (21-day-old) received ND injections before pilocarpine administration (400 mg/kg, s.c., pilocarpine group (P400)). The pretreatment with ND10 and ND30 prolonged the latencies of seizures and death on this seizure model. ND pretreatment in two doses decreased the levels of lipid peroxidation when compared to pilocarpine group. The P400 administration increased the striatal catalase activity. However, the administration of ND, in dose of 30 mg/kg, 30 min before pilocarpine, preserved catalase activity in normal levels. On the other hand, no change was detected in the animals treated with the dose of 10 mg/kg. Our results confirm the neuroprotective effect of ND on the seizures in young rats, suggesting that this drug acts positively on lipid peroxidation. Our observations shows that nimodipine cannot induces these effects via blockade of Ca(2+)-channel.
Collapse
Affiliation(s)
- Viviane S Nascimento
- Department of Physiology and Pharmacology, Laboratory of Neuropharmacology, School of Medicine, Federal University of Ceará. Rua Cel. Nunes de Melo 1127, 60431-270, Fortaleza, CE, Brazil
| | | | | | | | | | | | | |
Collapse
|
48
|
Aarts MM, Tymianski M. TRPMs and neuronal cell death. Pflugers Arch 2005; 451:243-9. [PMID: 16044308 DOI: 10.1007/s00424-005-1439-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Accepted: 04/09/2005] [Indexed: 12/21/2022]
Abstract
Death of CNS neurons during acute injury occurs as a result of a complex combination of excitotoxicity, necrosis, apoptosis, oedema and inflammatory reactions. Neuroprotection via glutamate receptor blockade or antioxidant or anti-inflammatory therapy have not proven effective in the clinical treatment of brain damage due to narrow therapeutic windows, poor pharmacokinetics or blockade of the signalling essential for normal excitatory neurotransmission and neuronal survival. Recent work in neuronal biochemistry, genomics and proteomics has increased understanding of the molecular organization of the excitatory synapse and the neuronal postsynaptic density. Transient receptor potential (TRP) channels are an exciting new family of cation channels that are highly expressed in the brain. Several members can be induced by oxidative stress and oxygen free radicals, both of which play important roles in neurodegeneration. Recent work has indicated that members of the melastatin subfamily (TRPM) of TRP proteins, particularly TRPM7 and TRPM2, may play key roles in neuronal death that is activated by oxidative stress and downstream from excitotoxic signal pathways. This discovery provides an exiting new avenue for research into the pathophysiology and treatment of acute neurodegeneration.
Collapse
Affiliation(s)
- Michelle M Aarts
- Applied and Interventional Research and Division of Neurosurgery, Toronto Western Research Institute, W4-325, 399 Bathurst Street, Toronto, ON M5T 2S8, Canada
| | | |
Collapse
|
49
|
Wainwright MS, Arteaga E, Fink R, Ravi K, Chace DH, Black SM. Tetrahydrobiopterin and nitric oxide synthase dimer levels are not changed following hypoxia-ischemia in the newborn rat. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2005; 156:183-92. [PMID: 16099305 DOI: 10.1016/j.devbrainres.2005.02.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Revised: 02/09/2005] [Accepted: 02/17/2005] [Indexed: 10/25/2022]
Abstract
The effect of hypoxia-ischemia on the nitric oxide synthase (NOS) cofactor tetrahydrobiopterin (BH4) and changes in the enzyme dimer state have not previously been studied. Cell-based studies have demonstrated the regulation of nitric oxide (NO) synthesis by intracellular BH4 levels. Activation of NOS requires two NOS polypeptides to form a homodimer. Dimerization results in the creation of high-affinity binding sites for BH4 and L-arginine. Our previous studies have indicated that nNOS activity falls 2 h post-hypoxia-ischemia in the immature rodent model. Thus, the objective of this study was to determine whether changes in nNOS dimeric state could be responsible for the decrease in nNOS activity. Using the immature rat model of HI in conjunction with LT-PAGE and Western blot analysis, we determined the effect of HI on NOS dimer state in hippocampus and cortex and the effects of pharmacologic modulation of NO levels during HI on dimer formation. Using high-performance liquid chromatography (HPLC) and electrospray tandem mass spectrometry (MS-MS), we measured BH4 and L-arginine levels respectively after HI under the same conditions. We found minimal or no changes in either BH4 levels or NOS dimer state at 2 h, 24 h and 7 day recovery from HI on postnatal day 7. In contrast, L-arginine levels were transiently increased in the hypoxic ischemic hemisphere. Thus, our data suggest that the previously described decrease in NOS activity after HI is not associated with depletion of the cofactor BH4, L-arginine substrate or changes in the NOS enzyme dimer state.
Collapse
Affiliation(s)
- Mark S Wainwright
- Department of Pediatrics, Divisions of Neurology, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
50
|
Dunlop J, Zaleska MM, Eliasof S, Moyer JA. Excitatory amino acid transporters as emerging targets for central nervous system therapeutics. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.3.4.543] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|