1
|
Khatibi VA, Rahdar M, Rezaei M, Davoudi S, Nazari M, Mohammadi M, Raoufy MR, Mirnajafi-Zadeh J, Hosseinmardi N, Behzadi G, Janahmadi M. The Glycolysis Inhibitor 2-Deoxy-D-Glucose Exerts Different Neuronal Effects at Circuit and Cellular Levels, Partially Reverses Behavioral Alterations and does not Prevent NADPH Diaphorase Activity Reduction in the Intrahippocampal Kainic Acid Model of Temporal Lobe Epilepsy. Neurochem Res 2023; 48:210-228. [PMID: 36064822 PMCID: PMC9444119 DOI: 10.1007/s11064-022-03740-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/31/2022] [Accepted: 08/27/2022] [Indexed: 01/11/2023]
Abstract
Temporal lobe epilepsy is the most drug-resistant type with the highest incidence among the other focal epilepsies. Metabolic manipulations are of great interest among others, glycolysis inhibitors like 2-deoxy D-glucose (2-DG) being the most promising intervention. Here, we sought to investigate the effects of 2-DG treatment on cellular and circuit level electrophysiological properties using patch-clamp and local field potentials recordings and behavioral alterations such as depression and anxiety behaviors, and changes in nitric oxide signaling in the intrahippocampal kainic acid model. We found that epileptic animals were less anxious, more depressed, with more locomotion activity. Interestingly, by masking the effect of increased locomotor activity on the parameters of the zero-maze test, no altered anxiety behavior was noted in epileptic animals. However, 2-DG could partially reverse the behavioral changes induced by kainic acid. The findings also showed that 2-DG treatment partially suppresses cellular level alterations while failing to reverse circuit-level changes resulting from kainic acid injection. Analysis of NADPH-diaphorase positive neurons in the CA1 area of the hippocampus revealed that the number of positive neurons was significantly reduced in dorsal CA1 of the epileptic animals and 2-DG treatment did not affect the diminishing effect of kainic acid on NADPH-d+ neurons in the CA1 area. In the control group receiving 2-DG, however, an augmented NADPH-d+ cell number was noted. These data suggest that 2-DG cannot suppress epileptiform activity at the circuit-level in this model of epilepsy and therefore, may fail to control the seizures in temporal lobe epilepsy cases.
Collapse
Affiliation(s)
- Vahid Ahli Khatibi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Shima Davoudi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Nazari
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Mohammad Mohammadi
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Mohammad Reza Raoufy
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Heylen L, Pham DH, De Meulemeester AS, Samarut É, Skiba A, Copmans D, Kazwiny Y, Vanden Berghe P, de Witte PAM, Siekierska A. Pericardial Injection of Kainic Acid Induces a Chronic Epileptic State in Larval Zebrafish. Front Mol Neurosci 2021; 14:753936. [PMID: 34720874 PMCID: PMC8551382 DOI: 10.3389/fnmol.2021.753936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a common disorder of the brain characterized by spontaneous recurrent seizures, which develop gradually during a process called epileptogenesis. The mechanistic processes underlying the changes of brain tissue and networks toward increased seizure susceptibility are not fully understood. In rodents, injection of kainic acid (KA) ultimately leads to the development of spontaneous epileptic seizures, reflecting similar neuropathological characteristics as seen in patients with temporal lobe epilepsy (TLE). Although this model has significantly contributed to increased knowledge of epileptogenesis, it is technically demanding, costly to operate and hence not suitable for high-throughput screening of anti-epileptic drugs (AEDs). Zebrafish, a vertebrate with complementary advantages to rodents, is an established animal model for epilepsy research. Here, we generated a novel KA-induced epilepsy model in zebrafish larvae that we functionally and pharmacologically validated. KA was administered by pericardial injection at an early zebrafish larval stage. The epileptic phenotype induced was examined by quantification of seizure-like behavior using automated video recording, and of epileptiform brain activity measured via local field potential (LFP) recordings. We also assessed GFP-labeled GABAergic and RFP-labeled glutamatergic neurons in double transgenic KA-injected zebrafish larvae, and examined the GABA and glutamate levels in the larval heads by liquid chromatography with tandem mass spectrometry detection (LC-MS/MS). Finally, KA-injected larvae were exposed to five commonly used AEDs by immersion for pharmacological characterization of the model. Shortly after injection, KA induced a massive damage and inflammation in the zebrafish brain and seizure-like locomotor behavior. An abnormal reorganization of brain circuits was observed, a decrease in both GABAergic and glutamatergic neuronal population and their associated neurotransmitters. Importantly, these changes were accompanied by spontaneous and continuous epileptiform brain discharges starting after a short latency period, as seen in KA rodent models and reminiscent of human pathology. Three out of five AEDs tested rescued LFP abnormalities but did not affect the seizure-like behavior. Taken together, for the first time we describe a chemically-induced larval zebrafish epilepsy model offering unique insights into studying epileptogenic processes in vivo and suitable for high-throughput AED screening purposes and rapid genetic investigations.
Collapse
Affiliation(s)
- Lise Heylen
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | - Duc-Hung Pham
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | | | - Éric Samarut
- Department of Neurosciences, Research Center of the University of Montreal Hospital Center, University of Montreal, Montreal, QC, Canada.,Modelis Inc., Montreal, QC, Canada
| | - Adrianna Skiba
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | - Daniëlle Copmans
- Laboratory for Molecular Biodiscovery, KU Leuven, Leuven, Belgium
| | - Youcef Kazwiny
- Laboratory for Enteric Neuroscience, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
3
|
Al Mughairbi F, Nawaz R, Khan F, Hassan A, Mahmood N, Ahmed HT, Alshamali A, Ahmed S, Bashir A. Neuroprotective effects of Bhilawanol and Anacardic acid during glutamate-induced neurotoxicity. Saudi Pharm J 2021; 29:1043-1049. [PMID: 34588850 PMCID: PMC8463467 DOI: 10.1016/j.jsps.2021.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/04/2021] [Indexed: 11/19/2022] Open
Abstract
Bhilawanol (Bh) and anacardic acid (AA) are two lipid-soluble compounds mostly found in the nut of Semecarpus anacardium (SA). This herb has many medicinal properties including enhancing learning and memory, yet its active compounds have not been studied for neuroprotective effects. We investigated the neuroprotective effects of Bh and AA against glutamate induced cell death in the adrenal pheochromocytoma cell line of rats (PC12 cells). Cell viability, toxicity and calcium influx were determined by MTT assay, LDH release assay and Fluo-3 imaging while apoptosis was assayed by caspase-3 and Bcl-2 gene expression. Our results showed that Bh and AA treatments significantly increased cell viability, reduced cell toxicity and calcium influx in PC12 cells in addition to suppressing the reactive oxygen species. Furthermore, AA treatment decreased caspase-3 expression level whereas both Bh and AA enhanced the expression of anti-apoptotic gene Bcl-2 in PC12 cells. Both compounds potently inhibited acetylcholinesterase enzyme (AChE) in a dose and time dependent manner. These findings suggest that the traditional use of SA may be explained on the basis of both Bh and AA showing neuroprotective potential due to their effects on enhancing cell viability, reducing cell toxicity most probably by reducing excessive calcium influx and suppression of ROS as well as by decreasing the expression of proapoptotic caspase 3 gene and increasing the expression of antiapoptotic gene Bcl2. Traditional use in enhancing learning and memory was justified in part by inhibition of AChE.
Collapse
Affiliation(s)
- Fadwa Al Mughairbi
- Dep. of Clinical Psychology, College of Medicines and Health Sciences, UAE University, Al Ain, United Arab Emirates
- Corresponding author at: Dep of Clinical Psychology, College of Medicine & Health Sciences, UAE University, Al Ain, United Arab Emirates.
| | - Rukhsana Nawaz
- Dep. of Clinical Psychology, College of Medicines and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Faisal Khan
- Panjwani Center for Molecular Medicine and Drug Research (PCMD), International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Amina Hassan
- Dep. of Clinical Psychology, College of Medicines and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Nailah Mahmood
- Dep. of Clinical Psychology, College of Medicines and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Heba Tageldeen Ahmed
- Dep. of Clinical Psychology, College of Medicines and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Alia Alshamali
- Dep. of Clinical Psychology, College of Medicines and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Sagheer Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Asma Bashir
- Endodontic Department, Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
4
|
The Kainic Acid Models of Temporal Lobe Epilepsy. eNeuro 2021; 8:ENEURO.0337-20.2021. [PMID: 33658312 PMCID: PMC8174050 DOI: 10.1523/eneuro.0337-20.2021] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/14/2021] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
Experimental models of epilepsy are useful to identify potential mechanisms of epileptogenesis, seizure genesis, comorbidities, and treatment efficacy. The kainic acid (KA) model is one of the most commonly used. Several modes of administration of KA exist, each producing different effects in a strain-, species-, gender-, and age-dependent manner. In this review, we discuss the advantages and limitations of the various forms of KA administration (systemic, intrahippocampal, and intranasal), as well as the histologic, electrophysiological, and behavioral outcomes in different strains and species. We attempt a personal perspective and discuss areas where work is needed. The diversity of KA models and their outcomes offers researchers a rich palette of phenotypes, which may be relevant to specific traits found in patients with temporal lobe epilepsy.
Collapse
|
5
|
High levels of 27-hydroxycholesterol results in synaptic plasticity alterations in the hippocampus. Sci Rep 2021; 11:3736. [PMID: 33580102 PMCID: PMC7881004 DOI: 10.1038/s41598-021-83008-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/25/2021] [Indexed: 12/16/2022] Open
Abstract
Alterations in brain cholesterol homeostasis in midlife are correlated with a higher risk of developing Alzheimer’s disease (AD). However, global cholesterol-lowering therapies have yielded mixed results when it comes to slowing down or preventing cognitive decline in AD. We used the transgenic mouse model Cyp27Tg, with systemically high levels of 27-hydroxycholesterol (27-OH) to examine long-term potentiation (LTP) in the hippocampal CA1 region, combined with dendritic spine reconstruction of CA1 pyramidal neurons to detect morphological and functional synaptic alterations induced by 27-OH high levels. Our results show that elevated 27-OH levels lead to enhanced LTP in the Schaffer collateral-CA1 synapses. This increase is correlated with abnormally large dendritic spines in the stratum radiatum. Using immunohistochemistry for synaptopodin (actin-binding protein involved in the recruitment of the spine apparatus), we found a significantly higher density of synaptopodin-positive puncta in CA1 in Cyp27Tg mice. We hypothesize that high 27-OH levels alter synaptic potentiation and could lead to dysfunction of fine-tuned processing of information in hippocampal circuits resulting in cognitive impairment. We suggest that these alterations could be detrimental for synaptic function and cognition later in life, representing a potential mechanism by which hypercholesterolemia could lead to alterations in memory function in neurodegenerative diseases.
Collapse
|
6
|
Micheli L, Di Cesare Mannelli L, Lucarini E, Parisio C, Toti A, Fiorentino B, Rigamonti MA, Calosi L, Ghelardini C. Intranasal Low-Dose Naltrexone Against Opioid Side Effects: A Preclinical Study. Front Pharmacol 2020; 11:576624. [PMID: 33071790 PMCID: PMC7531600 DOI: 10.3389/fphar.2020.576624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022] Open
Abstract
Opioids are broad spectrum analgesics that are an integral part of the therapeutic armamentarium to combat pain in the clinical practice. Unfortunately, together with analgesia, a number of adverse effects can occur such as nausea, vomiting, constipation, gastrointestinal alterations and cognitive impairments. Naltrexone is a competitive antagonist of opioid receptors commonly used to treat opioid addiction; its oral use against agonists side effects is limited by the decrease of opioids-therapeutic efficacy and own adverse effects. The intranasal delivery of naltrexone could offer a quick and effective achievement of CNS based on extracellular mechanisms including perineural and perivascular transport. The aim of the study was to test the efficacy of intranasal low-dose naltrexone in reducing intraperitoneal morphine and oxycodone side effects in rodents. In mice, 1 μg naltrexone intranasally administered 30 min before opioids reduced cognitive impairments and motor alteration induced by 10 mg kg-1 morphine and 60 mg kg-1 oxycodone in the Passive avoidance and Rota rod tests, respectively. Moreover, naltrexone rebalanced opioid-induced reduction of the intestinal transit and latency of feces expulsion as well as food intake inhibition. Importantly, 1 μg naltrexone instillation did not block analgesia as demonstrated by the Hot plate test. In rats, intranasal naltrexone counteracted the opioid-induced pica phenomenon related to emesis and increased water and palatable food intake. The effects were comparable to that achieved by metoclopramide used as reference drug. Treatments did not influence body weight. Lastly, the safety of the intranasal delivery has been checked by hematoxylin-eosin staining that did not show histological alterations of the nasal cavity. In conclusion, intranasal low-dose naltrexone counteracted morphine and oxycodone induced gastrointestinal and CNS side effects without impairing opioid analgesia. It is a candidate to be a valid clinical strategy deserving deep analysis.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | | | | | - Laura Calosi
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
7
|
Srivastava A, Liachenko S, Sarkar S, Paule M, Negi G, Pandey JP, Hanig JP. Quantitative Neurotoxicology: An Assessment of the Neurotoxic Profile of Kainic Acid in Sprague Dawley Rats. Int J Toxicol 2020; 39:294-306. [PMID: 32468881 DOI: 10.1177/1091581820928497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study consisted of a qualitative and quantitative assessment of neuropathological changes in kainic acid (KA)-treated adult male rats. Rats were administered a single 10 mg/kg intraperitoneal injection of KA or the same volume of saline and sacrificed 24 or 48 hours posttreatment. Brains were collected, sectioned coronally (∼ 81 slices), and stained with amino cupric silver to reveal degenerative changes. For qualitative assessment of neural degeneration, sectioned material was evaluated by a board-certified pathologist, and the level of degeneration was graded based upon a 4-point scale. For measurement of quantitative neural degeneration in response to KA treatment, the HALO digital image analysis software tool was used. Quantitative measurements of specific regions within the brain were obtained from silver-stained tissue sections with quantitation based on stain color and optical density. This quantitative evaluation method identified degeneration primarily in the cerebral cortex, septal nuclei, amygdala, olfactory bulb, hippocampus, thalamus, and hypothalamus. The KA-produced neuronal degeneration in the cortex was primarily in the piriform, insular, rhinal, and cingulate areas. In the hippocampus, the dentate gyrus was found to be the most affected area. Our findings indicate global neurotoxicity due to KA treatment. Certain brain structures exhibited more degeneration than others, reflecting differential sensitivity or vulnerability of neurons to KA.
Collapse
Affiliation(s)
| | - Serguei Liachenko
- National Center for Toxicological Research, NCTR/DNT, Jefferson, AR, USA
| | - Sumit Sarkar
- National Center for Toxicological Research, NCTR/DNT, Jefferson, AR, USA
| | - Merle Paule
- National Center for Toxicological Research, NCTR/DNT, Jefferson, AR, USA
| | - Geeta Negi
- US Food and Drug Administration, CDER/OPQ, Silver Spring, MD, USA
| | - Jai P Pandey
- US Food and Drug Administration, CDER/OPQ, Silver Spring, MD, USA
| | - Joseph P Hanig
- US Food and Drug Administration, CDER/OPQ, Silver Spring, MD, USA
| |
Collapse
|
8
|
Chaskiel L, Bristow AD, Bluthé RM, Dantzer R, Blomqvist A, Konsman JP. Interleukin-1 reduces food intake and body weight in rat by acting in the arcuate hypothalamus. Brain Behav Immun 2019; 81:560-573. [PMID: 31310797 DOI: 10.1016/j.bbi.2019.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022] Open
Abstract
A reduction in food intake is commonly observed after bacterial infection, a phenomenon that can be reproduced by peripheral administration of Gram-negative bacterial lipopolysaccharide (LPS) or interleukin-1beta (IL-1β), a pro-inflammatory cytokine released by LPS-activated macrophages. The arcuate nucleus of the hypothalamus (ARH) plays a major role in food intake regulation and expresses IL-1 type 1 receptor (IL-1R1) mRNA. In the present work, we tested the hypothesis that IL-1R1 expressing cells in the ARH mediate IL-1β and/or LPS-induced hypophagia in the rat. To do so, we developed an IL-1β-saporin conjugate, which eliminated IL-R1-expressing neurons in the hippocampus, and micro-injected it into the ARH prior to systemic IL-1β and LPS administration. ARH IL-1β-saporin injection resulted in loss of neuropeptide Y-containing cells and attenuated hypophagia and weight loss after intraperitoneal IL-1β, but not LPS, administration. In conclusion, the present study shows that ARH NPY-containing neurons express functional IL-1R1s that mediate peripheral IL-1β-, but not LPS-, induced hypophagia. Our present and previous findings indicate that the reduction of food intake after IL-1β and LPS are mediated by different neural pathways.
Collapse
Affiliation(s)
- Léa Chaskiel
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Adrian D Bristow
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK
| | - Rose-Marie Bluthé
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, The University of Texas, Houston, TX 770030, USA
| | - Anders Blomqvist
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Jan Pieter Konsman
- UMR CNRS 5287 Aquitaine Institute for Integrative and Cognitive Neuroscience, University of Bordeaux, 33076 Bordeaux, France.
| |
Collapse
|
9
|
Promising Neuroprotective Function for M2 Microglia in Kainic Acid-Induced Neurotoxicity Via the Down-Regulation of NF-κB and Caspase 3 Signaling Pathways. Neuroscience 2019; 406:86-96. [PMID: 30858108 DOI: 10.1016/j.neuroscience.2019.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/24/2022]
Abstract
Activated microglia have two functional states (M1 and M2) which play dual roles in neurodegenerative diseases. In the present study, we explored a possible neuroprotective function of M2 microglia against kainic acid (KA)-induced neurodegeneration in primary neurons co-cultured with different microglial populations. Neurons were isolated from the hippocampi and cortices of C57BL/6 embryos (embryonic day 16) and microglia were extracted from neonatal pups (postnatal days 0-2). Microglia were either unstimulated (M0-phenotype) or stimulated with lipopolysaccharide and interferon-γ to form the M1-phenotype, or with interleukin (IL)-4, IL-10, and transforming growth factor -β for the M2-phenotype. Neurons were co-cultured with each of the three microglial phenotypes and treated with KA for 24 h. Next, we analyzed the cell survival rate, nitric oxide (NO) levels, and lactate dehydrogenase production, cytokines levels, and expression of nuclear factor κB (NF-κB) and caspase 3 among the three groups before and after KA insult. Our results indicated that M2 microglia played a neuroprotective role in KA-induced neurotoxicity, as demonstrated by high neuronal survival as well as decreased production of NO and pro-inflammatory cytokines. In contrast, neurons co-cultured with M1 microglia exhibited the lowest survival rate as well as increased levels of NO and pro-inflammatory cytokines. Further, the expression of NF-κB and caspase 3 were significantly decreased in M2 microglia co-cultures compared to M1 or M0 microglia co-cultures after KA insult. Therefore, M2 microglia may exert a neuroprotective function in KA-induced neurotoxicity via the down-regulation of NF-κB and caspase 3 signaling pathways.
Collapse
|
10
|
Ghasemi-Kasman M, Baharvand H, Javan M. Enhanced neurogenesis in degenerated hippocampi following pretreatment with miR-302/367 expressing lentiviral vector in mice. Biomed Pharmacother 2017; 96:1222-1229. [PMID: 29174574 DOI: 10.1016/j.biopha.2017.11.094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/11/2017] [Accepted: 11/17/2017] [Indexed: 11/15/2022] Open
Abstract
Astrogliosis is the main landmark of neurodegenerative diseases. In vivo reprogramming of reactive astrocytes to functional neurons opened a new horizon in regenerative medicine. However there is little evidence that show possible application of in vivo reprogramming approaches for enhancement of neurogenesis. Cluster miR-302/367 showed high capability in cell reprogramming. Here we show that application of lentiviral particles expressing cluster miR-302/367 along with systemic valproate (VPA) enhanced the capability of mice brains for neurogenesis in CA3 area following kainic acid (KA) induced hippocampal neurodegeneration. Following pretreatment with miR-302/367 expressing viral particles and VPA, transduced cells showed neuroblast and mature neuron markers when neuronal loss was induced by KA. Comparing the neuron counts in CA3 region also showed that neurogenesis was increased in CA3 region in animals which were pretreated with miR-302/367 vector and VPA, only in injected side of the brain. Our data suggest that targeted application of miR-302/367 expressing vector may enhance the capacity of hippocampus and other brain structures for regeneration following neuronal loss.
Collapse
Affiliation(s)
- Maryam Ghasemi-Kasman
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
11
|
Pachenari N, Kiani S, Javan M. Inhibition of glycogen synthase kinase 3 increased subventricular zone stem cells proliferation. Biomed Pharmacother 2017; 93:1074-1082. [PMID: 28738501 DOI: 10.1016/j.biopha.2017.07.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/30/2017] [Accepted: 07/09/2017] [Indexed: 11/26/2022] Open
Abstract
The effects of Wnt signaling modifiers on cell proliferation, seem to be cell specific. Enhancing the proliferation of subventricular zone (SVZ) progenitors has been in the focus of research in recent years. Here we investigate the effect of CHIR99021, a Glycogen Synthase Kinase 3 (GSk-3) inhibitor, on SVZ progenitor's proliferation both in vivo and in vitro. Neural stem cells were extracted from the adult C57bl/6 by mincing and trypsin treatment followed by culturing in specific medium. Sphere cells formed within about 7-10days and were characterized by immunostaining. Number of spheres and their size was assessed following exposure to different concentration of CHIR99021 or vehicle. For in vivo studies, animals received intracerebroventricular (i.c.v.) injection of CHIR99021 or vehicle for four days. A subgroup of animals, after 4days treatment with CHIR99021 received intranasal kainic acid to induce local neurodegeneration in CA3 area of hippocampus. Inhibition of GSk-3 by CHIR99021 increased neural progenitor proliferation and the effect of CHIR99021 was long lasting so that the treated cells showed higher proliferation even after CHIR99021 removal. In vivo administration of CHIR99021 increased the number of neural progenitors at the rims of lateral ventricles especially when the treatment was followed by kainic acid administration which induces neural insult. Results showed that direct administration of CHIR99021 into the culture medium or animal brain increased the number of SVZ progenitors, especially when a neural insult was induced in the hippocampus.
Collapse
Affiliation(s)
- Narges Pachenari
- Department of Physiology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sahar Kiani
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
12
|
Glufosinate aerogenic exposure induces glutamate and IL-1 receptor dependent lung inflammation. Clin Sci (Lond) 2016; 130:1939-54. [PMID: 27549113 DOI: 10.1042/cs20160530] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/22/2016] [Indexed: 11/17/2022]
Abstract
Glufosinate-ammonium (GLA), the active component of an herbicide, is known to cause neurotoxicity. GLA shares structural analogy with glutamate. It is a powerful inhibitor of glutamine synthetase (GS) and may bind to glutamate receptors. Since these potentials targets of GLA are present in lung and immune cells, we asked whether airway exposure to GLA may cause lung inflammation in mice. A single GLA exposure (1 mg/kg) induced seizures and inflammatory cell recruitment in the broncho-alveolar space, and increased myeloperoxidase (MPO), inducible NO synthase (iNOS), interstitial inflammation and disruption of alveolar septae within 6-24 h. Interleukin 1β (IL-1β) was increased and lung inflammation depended on IL-1 receptor 1 (IL-1R1). We demonstrate that glutamate receptor pathway is central, since the N-methyl-D-aspartate (NMDA) receptor inhibitor MK-801 prevented GLA-induced lung inflammation. Chronic exposure (0.2 mg/kg 3× per week for 4 weeks) caused moderate lung inflammation and enhanced airway hyperreactivity with significant increased airway resistance. In conclusion, GLA aerosol exposure causes glutamate signalling and IL-1R-dependent pulmonary inflammation with airway hyperreactivity in mice.
Collapse
|
13
|
Najyb O, Do Carmo S, Alikashani A, Rassart E. Apolipoprotein D Overexpression Protects Against Kainate-Induced Neurotoxicity in Mice. Mol Neurobiol 2016; 54:3948-3963. [PMID: 27271124 PMCID: PMC7091089 DOI: 10.1007/s12035-016-9920-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 05/03/2016] [Indexed: 01/23/2023]
Abstract
Excitotoxicity due to the excessive activation of glutamatergic receptors leads to neuronal dysfunction and death. Excitotoxicity has been implicated in the pathogenesis of a myriad of neurodegenerative diseases with distinct etiologies such as Alzheimer's and Parkinson's. Numerous studies link apolipoprotein D (apoD), a secreted glycoprotein highly expressed in the central nervous system (CNS), to maintain and protect neurons in various mouse models of acute stress and neurodegeneration. Here, we used a mouse model overexpressing human apoD in neurons (H-apoD Tg) to test the neuroprotective effects of apoD in the kainic acid (KA)-lesioned hippocampus. Our results show that apoD overexpression in H-apoD Tg mice induces an increased resistance to KA-induced seizures, significantly attenuates inflammatory responses and confers protection against KA-induced cell apoptosis in the hippocampus. The apoD-mediated protection against KA-induced toxicity is imputable in part to increased plasma membrane Ca2+ ATPase type 2 expression (1.7-fold), decreased N-methyl-D-aspartate receptor (NMDAR) subunit NR2B levels (30 %) and lipid metabolism alterations. Indeed, we demonstrate that apoD can attenuate intracellular cholesterol content in primary hippocampal neurons and in brain of H-apoD Tg mice. In addition, apoD can be internalised by neurons and this internalisation is accentuated in ageing and injury conditions. Our results provide additional mechanistic information on the apoD-mediated neuroprotection in neurodegenerative conditions.
Collapse
Affiliation(s)
- Ouafa Najyb
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada
| | - Sonia Do Carmo
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Azadeh Alikashani
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada
| | - Eric Rassart
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada.
| |
Collapse
|
14
|
Sabilallah M, Fontanaud P, Linck N, Boussadia B, Peyroutou R, Lasgouzes T, Rassendren FA, Marchi N, Hirbec HE. Evidence for Status Epilepticus and Pro-Inflammatory Changes after Intranasal Kainic Acid Administration in Mice. PLoS One 2016; 11:e0150793. [PMID: 26963100 PMCID: PMC4786335 DOI: 10.1371/journal.pone.0150793] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/21/2016] [Indexed: 12/04/2022] Open
Abstract
Kainic acid (KA) is routinely used to elicit status epilepticus (SE) and epileptogenesis. Among the available KA administration protocols, intranasal instillation (IN) remains understudied. Dosages of KA were instilled IN in mice. Racine Scale and Video-EEG were used to assess and quantify SE onset. Time spent in SE and spike activity was quantified for each animal and confirmed by power spectrum analysis. Immunohistochemistry and qPCR were performed to define brain inflammation occurring after SE, including activated microglial phenotypes. Long term video-EEG recording was also performed. Titration of IN KA showed that a dose of 30 mg/kg was associated with low mortality while eliciting SE. IN KA provoked at least one behavioral and electrographic SE in the majority of the mice (>90%). Behavioral and EEG SE were accompanied by a rapid and persistent microglial-astrocytic cell activation and hippocampal neurodegeneration. Specifically, microglial modifications involved both pro- (M1) and anti-inflammatory (M2) genes. Our initial long-term video-EEG exploration conducted using a small cohort of mice indicated the appearance of spike activity or SE. Our study demonstrated that induction of SE is attainable using IN KA in mice. Typical pro-inflammatory brain changes were observed in this model after SE, supporting disease pathophysiology. Our results are in favor of the further development of IN KA as a means to study seizure disorders. A possibility for tailoring this model to drug testing or to study mechanisms of disease is offered.
Collapse
Affiliation(s)
- Mounira Sabilallah
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Labex ICST, Montpellier, France
| | - Pierre Fontanaud
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Plateforme Imagerie du Petit Animal Montpellier, Biocampus, Montpellier, France
| | - Nathalie Linck
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Labex ICST, Montpellier, France
| | - Badreddine Boussadia
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
| | - Ronan Peyroutou
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Labex ICST, Montpellier, France
| | - Thibault Lasgouzes
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Labex ICST, Montpellier, France
| | - François A. Rassendren
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Labex ICST, Montpellier, France
| | - Nicola Marchi
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
| | - Helene E. Hirbec
- CNRS, UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
- INSERM, U1191, Montpellier, France
- Université de Montpellier, UMR5203, Montpellier, France
- Labex ICST, Montpellier, France
| |
Collapse
|
15
|
Immediate Epileptogenesis after Kainate-Induced Status Epilepticus in C57BL/6J Mice: Evidence from Long Term Continuous Video-EEG Telemetry. PLoS One 2015; 10:e0131705. [PMID: 26161754 PMCID: PMC4498886 DOI: 10.1371/journal.pone.0131705] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/04/2015] [Indexed: 11/19/2022] Open
Abstract
The C57BL/6J mouse as a model of seizure/epilepsy is challenging due to high mortality and huge variability in response to kainate. We have recently demonstrated that repeated administration of a low dose of kainate by intraperitoneal route can induce severe status epilepticus (SE) with 94% survival rate. In the present study, based on continuous video-EEG recording for 4-18 weeks from epidurally implanted electrodes on the cortex, we demonstrate that this method also induces immediate epileptogenesis (<1-5 days post-SE). This finding was based on identification of two types of spontaneous recurrent seizures; behavioral convulsive seizures (CS) and electrographic nonconvulsive seizures (NCS). The identification of the spontaneous CS, stage 3-5 types, was based on the behaviors (video) that were associated with the EEG characteristics (stage 3-5 epileptiform spikes), the power spectrum, and the activity counts. The electrographic NCS identification was based on the stage 1-2 epileptiform spike clusters on the EEG and their associated power spectrum. Severe SE induced immediate epileptogenesis in all the mice. The maximum numbers of spontaneous CS were observed during the first 4-6 weeks of the SE and they decreased thereafter. Mild SE also induced immediate epileptogenesis in some mice but the CS were less frequent. In both the severe and the mild SE groups, the spontaneous electrographic NCS persisted throughout the 18 weeks observation period, and therefore this could serve as a chronic model for complex seizures. However, unlike rat kainate models, the C57BL/6J mouse kainate model is a unique regressive CS model of epilepsy. Further studies are required to understand the mechanism of recovery from spontaneous CS in this model, which could reveal novel therapeutic targets for epilepsy.
Collapse
|
16
|
Advantages of repeated low dose against single high dose of kainate in C57BL/6J mouse model of status epilepticus: behavioral and electroencephalographic studies. PLoS One 2014; 9:e96622. [PMID: 24802808 PMCID: PMC4011859 DOI: 10.1371/journal.pone.0096622] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/09/2014] [Indexed: 11/19/2022] Open
Abstract
A refined kainate (KA) C57BL/6J mouse model of status epilepticus (SE) using a repeated low dose (RLD) of KA (5 mg/kg, intraperitoneal; at 30 min intervals) was compared with the established single high dose (SHD) of KA (20 mg/kg, intraperitoneal) model. In the RLD group, increased duration of convulsive motor seizures (CMS, Racine scale stage ≥3) with a significant reduction in mortality from 21% to 6% and decreased variability in seizure severity between animals/batches were observed when compared to the SHD group. There was a significant increase in the percentage of animals that reached stage-5 seizures (65% versus 96%) in the RLD group. Integrated real-time video-EEG analysis of both groups, using NeuroScore software, revealed stage-specific spikes and power spectral density characteristics. When the seizures progressed from non-convulsive seizures (NCS, stage 1-2) to CMS (stage 3-5), the delta power decreased which was followed by an increase in gamma and beta power. A transient increase in alpha and sigma power marked the transition from NCS to CMS with characteristic 'high frequency trigger' spikes on the EEG, which had no behavioral expression. During SE the spike rate was higher in the RLD group than in the SHD group. Overall these results confirm that RLD of KA is a more robust and consistent mouse model of SE than the SHD of KA mouse model.
Collapse
|
17
|
Zhang XM, Mao XJ, Zhang HL, Zheng XY, Pham T, Adem A, Winblad B, Mix E, Zhu J. Overexpression of apolipoprotein E4 increases kainic-acid-induced hippocampal neurodegeneration. Exp Neurol 2011; 233:323-32. [PMID: 22079154 DOI: 10.1016/j.expneurol.2011.10.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 10/06/2011] [Accepted: 10/28/2011] [Indexed: 12/25/2022]
Abstract
Apolipoprotein E (apoE) has an intricate biological function in modulating immune responses and apoE isoforms exhibit diverse effects on neurodegenerative and neuroinflammatory disorders. In the present study, we investigated the individual roles of apoE isoforms in the kainic acid (KA)-induced hippocampal neurodegeneration with focus on immune response and microglia functions. ApoE2, 3 and 4 transgenic mice as well as wild-type (WT) mice were treated with KA by intranasal route. ApoE4 overexpressing mice revealed several peculiarities as compared with other transgenic mice and WT mice, i.e. (1) they had more severe KA-induced seizures than apoE2 and 3 mice, (2) they exhibited neuron loss in hippocampus that was higher than in apoE2, 3 and WT mice, (3) KA administration resulted in higher counts of their head drops in the cross-area of elevated plus-maze, (4) they showed lower KA-induced rearing activity than apoE2 mice in the open-field test, (5) their KA-induced microglial expression of MHC-II and CD86 was elevated compared to apoE3 mice, (6) the KA-induced increase of microglial iNOS was higher than that in the other groups of mice, and (7) the TNF-α and IL-6 expression was decreased 7 days after KA application compared to untreated mice and mice treated 1 day with KA. However, the signaling pathway of NFκB or Akt seemed not to be involved in apoE-isoform dependent susceptibility to KA-induced neurotoxicity. In conclusion, over-expression of apoE4 deteriorated KA-induced hippocampal neurodegeneration in C57BL/6 mice, which might result from a higher up-regulation of microglia activation compared to apoE2 and 3 transgenic mice and WT mice.
Collapse
Affiliation(s)
- Xing-Mei Zhang
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kainic acid-induced neurodegenerative model: potentials and limitations. J Biomed Biotechnol 2010; 2011:457079. [PMID: 21127706 PMCID: PMC2992819 DOI: 10.1155/2011/457079] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 10/25/2010] [Indexed: 01/03/2023] Open
Abstract
Excitotoxicity is considered to be an important mechanism involved in various neurodegenerative diseases in the central nervous system (CNS) such as Alzheimer's disease (AD). However, the mechanism by which excitotoxicity is implicated in neurodegenerative disorders remains unclear. Kainic acid (KA) is an epileptogenic and neuroexcitotoxic agent by acting on specific kainate receptors (KARs) in the CNS. KA has been extensively used as a specific agonist for ionotrophic glutamate receptors (iGluRs), for example, KARs, to mimic glutamate excitotoxicity in neurodegenerative models as well as to distinguish other iGluRs such as α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptors and N-methyl-D-aspartate receptors. Given the current knowledge of excitotoxicity in neurodegeneration, interventions targeted at modulating excitotoxicity are promising in terms of dealing with neurodegenerative disorders. This paper summarizes the up-to-date knowledge of neurodegenerative studies based on KA-induced animal model, with emphasis on its potentials and limitations.
Collapse
|
19
|
Serrano-Pérez MC, Martín ED, Vaquero CF, Azcoitia I, Calvo S, Cano E, Tranque P. Response of transcription factor NFATc3 to excitotoxic and traumatic brain insults: Identification of a subpopulation of reactive astrocytes. Glia 2010; 59:94-107. [DOI: 10.1002/glia.21079] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 08/23/2010] [Indexed: 01/21/2023]
|
20
|
Sharma S, Haselton J, Rakoczy S, Branshaw S, Brown-Borg HM. Spatial memory is enhanced in long-living Ames dwarf mice and maintained following kainic acid induced neurodegeneration. Mech Ageing Dev 2010; 131:422-35. [PMID: 20561541 DOI: 10.1016/j.mad.2010.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 05/18/2010] [Accepted: 06/05/2010] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Age associated cognitive impairment is associated with low levels of IGF-1, oxidative stress, and neuronal loss in the hippocampus. Ames dwarf mice are long-lived animals that exhibit peripheral IGF-1 deficiency. Hippocampal-based spatial memory (a homolog of cognitive function) has not been evaluated in these long-living mice. MATERIALS AND METHODS We evaluated the hippocampal-based spatial memory in 3-, 12- and 24-month-old Ames dwarf and wild type mice using the Barnes maze and the T-maze. We also examined the effect of a hippocampal-specific toxin, kainic acid (KA), on spatial memory to determine whether Ames mice were resistant to the cognitive impairment induced by this compound. RESULTS We found that Ames dwarf mice exhibit enhanced learning, making fewer errors and using less time to solve both the Barnes and T-mazes. Dwarf mice also have significantly better short-term memory as compared to wild type mice. Both genotypes exhibited neuronal loss in the CA1 and CA3 areas of the hippocampus following KA, but Ames dwarf mice retained their spatial memory. DISCUSSION Our results show that Ames dwarf mice retained their spatial memory despite neurodegeneration when compared to wild type mice at an "equiseizure" dose of KA.
Collapse
Affiliation(s)
- Sunita Sharma
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | | | | | | | | |
Collapse
|
21
|
Zhang XM, Jin T, Quezada HC, Mix E, Winblad B, Zhu J. Kainic acid-induced microglial activation is attenuated in aged interleukin-18 deficient mice. J Neuroinflammation 2010; 7:26. [PMID: 20398244 PMCID: PMC2865455 DOI: 10.1186/1742-2094-7-26] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 04/14/2010] [Indexed: 12/28/2022] Open
Abstract
Background Previously, we found that interleukin (IL)-18 deficiency aggravates kainic acid (KA)-induced hippocampal neurodegeneration in young C57BL/6 mice due to an over-compensation by IL-12. Additionally, IL-18 participates in fundamental inflammatory processes that increase during aging. In the present study, we were interested in the role of IL-18 in KA-induced neurodegeneration in aged female C57BL/6 mice. Methods Fifteen aged female IL-18 knockout (KO) and 15 age-matched wild-type (WT) mice (18 to 19 months old) were treated with KA at a dose of 25 mg/kg body weight intranasally. Seizure activities and behavioral changes were rated using a 6-point scoring system and open-field test, respectively. Seven days after KA treatment, degenerating neurons were detected by Nissl's method and Fluoro-Jade B staining; and microglial activation was analyzed by immunohistochemistry and flow cytometry. Results Aged female IL-18 KO and WT mice showed similar responses to treatment with KA as demonstrated by comparable seizure activities, behavioral changes and neuronal cell death. However, aged female IL-18 KO mice failed to exhibit the strong microglial activation shown in WT mice. Interestingly, even though the number of activated microglia was less in KA-treated IL-18 KO mice than in KA-treated WT mice, the proportion of microglia that expressed the cytokines tumor necrosis factor (TNF)-α, IL-6 and IL-10 was higher in KA-treated IL-18 KO mice. Conclusion Deficiency of IL-18 attenuates microglial activation after KA-induced excitotoxicity in aged brain, while the net effects of IL-18 deficiency are balanced by the enhancement of other cytokines, such as TNF-α, IL-6 and IL-10.
Collapse
Affiliation(s)
- Xing-Mei Zhang
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
22
|
Spigolon G, Veronesi C, Bonny C, Vercelli A. c-Jun N-terminal kinase signaling pathway in excitotoxic cell death following kainic acid-induced status epilepticus. Eur J Neurosci 2010; 31:1261-72. [DOI: 10.1111/j.1460-9568.2010.07158.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
23
|
Wang CK, Chen LG, Wen CL, Hou WC, Hung LF, Yen SJ, Shen YJ, Lin SY, Liang YC. Neuroprotective Activity of Vitis thunbergii var. taiwaniana Extracts In Vitro and In Vivo. J Med Food 2010; 13:170-8. [DOI: 10.1089/jmf.2009.1162] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Chung-Kwe Wang
- Department of Primary Care Medicine, Taipei Medical University Hospital
- Department of Internal Medicine, Renai Branch, Taipei City Hospital
| | - Lih-Geeng Chen
- Graduate Institute of Biomedical and Biopharmaceutical Sciences, College of Life Sciences, National Chiayi University, Chiayi
| | - Chi-Luan Wen
- Taiwan Seed Improvement and Propagation Station, Council of Agriculture
- Graduate Institute of Chinese Pharmaceutical Science, China Medical University, Taichung, Taiwan
| | - Wen-Chi Hou
- Graduate Institute of Pharmacognosy Science, College of Pharmacy, College of Medicine, Taipei Medical University, Taipei
| | - Ling-Fang Hung
- School of Medical Laboratory Science and Biotechnology, College of Medicine, Taipei Medical University, Taipei
| | - Shish-Jung Yen
- Medical Laboratory (Yang Ming Branch), Taipei City Hospital
| | - Yi-Jyun Shen
- School of Medical Laboratory Science and Biotechnology, College of Medicine, Taipei Medical University, Taipei
| | - Shyr-Yi Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital
| | - Yu-Chih Liang
- School of Medical Laboratory Science and Biotechnology, College of Medicine, Taipei Medical University, Taipei
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital
| |
Collapse
|
24
|
Lu MO, Zhang XM, Mix E, Quezada HC, Jin T, Zhu J, Adem A. TNF-alpha receptor 1 deficiency enhances kainic acid-induced hippocampal injury in mice. J Neurosci Res 2008; 86:1608-14. [PMID: 18189316 DOI: 10.1002/jnr.21600] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The exact role of TNF-alpha in excitotoxic neurodegeneration of the brain is unclear. To address this issue, the kainic acid (KA)-induced hippocampal injury model, a well-characterized model of human neurodegenerative diseases, was used in TNF-alpha receptor 1 (TNFR1)-knockout (TNFR1-/-) mice in the present study. After nasal application of a single dose of 40 mg of KA per kilogram body weight, TNFR1-/- mice showed significantly more severe seizures than the wild-type mice. In addition, obvious neurodegeneration, enhanced microglia activation, and astrogliosis in the hippocampus, as well as increased locomotor activity, were found in TNFR1-/- mice compared with the wild-type controls 8 days after KA delivery. Moreover, CC chemokine receptor 3 expression on activated microglia was increased 3 days after KA treatment in TNFR1-/- mice, as measured by flow cytometry. These data suggest that TNF-alpha may play a protective role through TNFR1 signaling.
Collapse
Affiliation(s)
- Ming-Ou Lu
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
25
|
Zhang XM, Zhu SW, Duan RS, Mohammed AH, Winblad B, Zhu J. Gender differences in susceptibility to kainic acid-induced neurodegeneration in aged C57BL/6 mice. Neurotoxicology 2008; 29:406-12. [PMID: 18342945 DOI: 10.1016/j.neuro.2008.01.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 01/10/2008] [Accepted: 01/29/2008] [Indexed: 11/15/2022]
Abstract
Some epidemiological studies concerning gender differences in Alzheimer's disease (AD) support the higher prevalence and incidence of AD in women, while most studies using animal models of aging have included only male subjects. It is still uncommon for aged males and females to be compared in the same study. In the present study, we investigated how age and gender influence the excitotoxic neurodegeneration by treating C57BL/6 mice (aged females and males as well as adult females and males) with kainic acid (KA) intranasally. Clinical signs, behavioural changes, pathological changes and astrocyte proliferation were tested; and the levels of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) were measured after KA treatment. The results showed that aged female mice were more sensitive to KA-induced excitotoxicity as demonstrated by severer seizure activity, increased locomotion and rearing in open-field test, prominent hippocampal neuronal damage, enhanced astrocyte proliferation compared with aged males, adult females and adult male mice. In addition, higher BDNF level in hippocampus of aged female mice was observed. These results denote the disparity of aging and gender in KA-induced hippocampal neurodegeneration and aged female mice are more sensitive to the excitotoxicity.
Collapse
Affiliation(s)
- Xing-Mei Zhang
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Scalia J, Lisanby SH, Dwork AJ, Johnson JE, Bernhardt ER, Arango V, McCall WV. Neuropathologic examination after 91 ECT treatments in a 92-year-old woman with late-onset depression. J ECT 2007; 23:96-8. [PMID: 17548979 DOI: 10.1097/yct.0b013e31804bb99d] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Whereas pathological seizure states, such as temporal lobe epilepsy, are commonly associated with cell loss and glial scarring in the hippocampus, seizures induced via electroconvulsive therapy (ECT) have not been associated with histological evidence of neuronal damage. We present a case report including the late-life medical history and postmortem histology of an elderly woman with major depression who received 91 sessions of ECT during the last 22 years of her life. Given the large number of ECT sessions, and her advanced age, this case provides a strong test of whether ECT causes detectable evidence of neuronal damage. We examined the gross morphology of the hippocampus, hippocampal cytoarchitecture, and measures of neuropathology. We found no pathological changes that could be attributed to ECT. Only expected, age-related features were present. Corpora amylacea and rare neurofibrillary tangles were evident, but we failed to detect any obvious evidence of cell loss or gliosis. Cognition in this patient was intact as indicated by a perfect score on a Mini-Mental Status Examination administered 6 days before death at the age of 92. This case adds to the considerable evidence for the safety of ECT.
Collapse
Affiliation(s)
- Jason Scalia
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Benkovic SA, O'Callaghan JP, Miller DB. Regional neuropathology following kainic acid intoxication in adult and aged C57BL/6J mice. Brain Res 2006; 1070:215-31. [PMID: 16403473 DOI: 10.1016/j.brainres.2005.11.065] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 11/03/2005] [Accepted: 11/06/2005] [Indexed: 01/05/2023]
Abstract
We evaluated regional neuropathological changes in adult and aged male mice treated systemically with kainic acid (KA) in a strain reported to be resistant to excitotoxic neuronal damage, C57BL/6. KA was administered in a single intraperitoneal injection. Adult animals were dosed with 35 mg/kg KA, while aged animals received a dose of 20 mg/kg in order to prevent excessive mortality. At time-points ranging from 12 h to 7 days post-treatment, animals were sacrificed and prepared for histological evaluation utilizing the cupric-silver neurodegeneration stain, immunohistochemistry for GFAP and IgG, and lectin staining. In animals of both ages, KA produced argyrophilia in neurons throughout cortex, hippocampus, thalamus, and amygdala. Semi-quantitative analysis of neuropathology revealed a similar magnitude of damage in animals of both ages, even though aged animals received less toxicant. Additional animals were evaluated for KA-induced reactive gliosis, assayed by an ELISA for GFAP, which revealed a 2-fold elevation in protein levels in adult mice, and a 2.5-fold elevation in aged animals. Histochemical evaluation of GFAP and lectin staining revealed activation of astrocytes and microglia in regions with corresponding argyrophilia. IgG immunostaining revealed a KA-induced breach of the blood-brain barrier in animals of both ages. Our data indicate widespread neurotoxicity following kainic acid treatment in C57BL/6J mice, and reveal increased sensitivity to this excitotoxicant in aged animals.
Collapse
Affiliation(s)
- Stanley Anthony Benkovic
- Toxicology and Molecular Biology Branch, Centers for Disease Control and Prevention-National Institute for Occupational, Safety and Health, Mailstop 3014, 1095 Willowdale Road, Morgantown, WV 26505, USA
| | | | | |
Collapse
|
29
|
Osterweil E, Wells DG, Mooseker MS. A role for myosin VI in postsynaptic structure and glutamate receptor endocytosis. ACTA ACUST UNITED AC 2005; 168:329-38. [PMID: 15657400 PMCID: PMC2171578 DOI: 10.1083/jcb.200410091] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myosin VI (Myo6) is an actin-based motor protein implicated in clathrin-mediated endocytosis in nonneuronal cells, though little is known about its function in the nervous system. Here, we find that Myo6 is highly expressed throughout the brain, localized to synapses, and enriched at the postsynaptic density. Myo6-deficient (Snell's waltzer; sv/sv) hippocampus exhibits a decrease in synapse number, abnormally short dendritic spines, and profound astrogliosis. Similarly, cultured sv/sv hippocampal neurons display decreased numbers of synapses and dendritic spines, and dominant-negative disruption of Myo6 in wild-type hippocampal neurons induces synapse loss. Importantly, we find that sv/sv hippocampal neurons display a significant deficit in the stimulation-induced internalization of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid-type glutamate receptors (AMPARs), and that Myo6 exists in a complex with the AMPAR, AP-2, and SAP97 in brain. These results suggest that Myo6 plays a role in the clathrin-mediated endocytosis of AMPARs, and that its loss leads to alterations in synaptic structure and astrogliosis.
Collapse
Affiliation(s)
- Emily Osterweil
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA.
| | | | | |
Collapse
|
30
|
Chen Z, Duan RS, Quezada HC, Mix E, Nennesmo I, Adem A, Winblad B, Zhu J. Increased microglial activation and astrogliosis after intranasal administration of kainic acid in C57BL/6 mice. ACTA ACUST UNITED AC 2005; 62:207-18. [PMID: 15459893 DOI: 10.1002/neu.20099] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Glutamate excitotoxicity plays a key role in inducing neuronal cell death in many neurological diseases. In mice, intranasal administration of kainic acid (KA), an analogue of the excitotoxin glutamate, results in hippocampal cell death and provides a well-characterized model for studies of human neurodegenerative diseases. In this study, we describe neurodegeneration and gliosis following intranasal administration of KA in C57BL/6 mice. By using Nissl's staining, neurodegeneration was found in area CA3 of hippocampus, and neuronal apoptosis was demonstrated by enhanced FAS(CD95/APO-1) expression detected by immunohistochemistry and Western blotting. Astrogliosis was exhibited by increased glial fibrillary acidic protein (GFAP) expression in the hippocampus and cortex. We also studied the profile of molecular expression on microglia in C57BL/6 mice. One and 3 days after KA administration, CD45, F4/80, CD86, MHCII, iNOS but not CD40 expression was enhanced or induced on microglia. In summary, KA administration results in an early microglial activation and a prolonged astrogliosis in C57BL/6 mice.
Collapse
Affiliation(s)
- Zhiguo Chen
- Division of Experimental Geriatrics, Department of Neurotec, Karolinska Institute, Karolinska University Hospital, Stockholm 14186, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Gahring LC, Persiyanov K, Days EL, Rogers SW. Age-related loss of neuronal nicotinic receptor expression in the aging mouse hippocampus corresponds with cyclooxygenase-2 and PPAR? expression and is altered by long-term NS398 administration. ACTA ACUST UNITED AC 2005; 62:453-68. [PMID: 15551346 DOI: 10.1002/neu.20106] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Age-related changes in the mammalian dorsal hippocampus are associated with diminished expression of neuronal nicotinic acetylcholine receptors (nAChR), which is particularly severe in pathologies such as those associated with dementias, including Alzheimer's disease. Because the mouse is a useful model for age-related decline in nAChR expression in the basal forebrain and limbic system, we used immunohistochemistry to examine the influence of long-term (12-month) oral administration of nicotine and/or the cyclooxygenase-2 (COX-2) preferring non-steroidal anti-inflammatory drug (NSAID) NS398 on nAChR alpha4, alpha5, alpha7, and beta4 expression in the C57BL/6 mouse. Inhibitory neurons of the dorsal hippocampus that express nAChRs also constitutively express COX-2 and the peroxisome proliferator-antagonist receptor subtype gamma-2 (PPAR gamma2) which is also a target of NS398. Administration of NS398 correlated with retention of nAChR alpha4 and to a lesser extent nAChR beta4, but not nAChR alpha5 or alpha7, but nicotine exhibited no similar effect. Nicotine and NS398 co-administration abolished the NS398-related effect on nAChR alpha4 retention. These results provide evidence that the interaction during aging between oral administration of nicotine and NSAIDs are not straightforward and could even be antagonistic when combined.
Collapse
Affiliation(s)
- Lorise C Gahring
- Salt Lake City VA-Geriatrics Research, Education and Clinical Center, Salt Lake City, Utah, USA
| | | | | | | |
Collapse
|
32
|
Chen Z, Duan RS, Concha QH, Wu Q, Mix E, Winblad B, Ljunggren HG, Zhu J. IL-12p35 deficiency alleviates kainic acid-induced hippocampal neurodegeneration in C57BL/6 mice. Neurobiol Dis 2004; 17:171-8. [PMID: 15474355 DOI: 10.1016/j.nbd.2004.07.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Revised: 06/23/2004] [Accepted: 07/30/2004] [Indexed: 11/29/2022] Open
Abstract
The role of IL-12 in excitotoxic neurodegeneration of brain is largely unknown. To address this issue, we used the model of kainic acid (KA)-induced hippocampal injury in IL-12p35 knockout (KO) mice, a well-characterized model for human neurodegenerative diseases. After KA treatment, hippocampal neurodegeneration was significantly less severe in the IL-12p35 KO mice than in wild-type mice as demonstrated by reduced pathological changes and astrogliosis. One day after KA treatment, levels of F4/80 and CD86 expression on microglia were significantly lower in IL-12p35 KO mice than in wild-type mice analyzed by flow cytometry, indicating that IL-12p35 deficiency resulted in lower levels of microglial activation. Five days after KA treatment, CD86 expression on microglia of wild-type mice was still higher, whereas F4/80 expression in wild-type mice decreased and was similar to that in IL-12p35 KO mice. Because microglial activation is necessary for KA-induced neurodegeneration, the lower level of microglial activation in the absence of IL-12p35 may alleviate hippocampal injury in KO mice. In summary, this study indicates that IL-12 may play a critical role in excitotoxin-induced brain injury.
Collapse
Affiliation(s)
- Zhiguo Chen
- Division of Experimental Geriatrics, Department of Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Watson GS, Craft S. Modulation of memory by insulin and glucose: neuropsychological observations in Alzheimer's disease. Eur J Pharmacol 2004; 490:97-113. [PMID: 15094077 DOI: 10.1016/j.ejphar.2004.02.048] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2004] [Indexed: 12/20/2022]
Abstract
Converging evidence has identified a potential association among Alzheimer's disease, glucose metabolism, insulin activity, and memory. Notably, type 2 diabetes, which is characterized by insulin resistance, may modulate the risk of Alzheimer's disease, and patients with Alzheimer's disease may have a greater risk for glucoregulatory impairments than do healthy older adults. In animal studies, it has been shown that raising blood glucose levels acutely can facilitate memory, in part, by increasing cholinergic activity, which is greatly diminished in patients with Alzheimer's disease. Other studies have confirmed that glucose administration can facilitate memory in healthy humans and in patients with Alzheimer's disease. Interestingly, glucose effects on memory appear to be modulated by insulin sensitivity (efficiency of insulin-mediated glucose disposal). Of course, the acute effects of glucose administration should be distinguished from the effects of chronic hyperglycemia (diabetes), which has been associated with cognitive impairments, at least in older adults. The relationship of insulin and memory has been more difficult to characterize. In animals, systemic insulin administration has been associated with memory deficits, likely due, in part, to hypoglycemia that occurs when exogenous insulin is not supplemented with glucose to maintain euglycemia. In healthy adults and patients with Alzheimer's disease, raising plasma insulin levels while maintaining euglycemia can improve memory; however, raising plasma glucose while suppressing endogenous insulin secretion may not improve memory, suggesting that adequate levels of insulin and glucose are necessary for memory facilitation. Clinical studies have corroborated findings that patients with Alzheimer's disease are more likely than healthy older adults to have reduced insulin sensitivity, and further suggest that apolipoprotein E genotype may modulate the effects of insulin on glucose disposal, memory facilitation, and amyloid precursor protein processing. Collectively, these findings support an association among Alzheimer's disease, impaired glucose metabolism, and reduced insulin sensitivity.
Collapse
Affiliation(s)
- G Stennis Watson
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Medical Center, 1660 South Columbian Way, Seattle, WA 98108, USA
| | | |
Collapse
|
34
|
Kitao Y, Hashimoto K, Matsuyama T, Iso H, Tamatani T, Hori O, Stern DM, Kano M, Ozawa K, Ogawa S. ORP150/HSP12A regulates Purkinje cell survival: a role for endoplasmic reticulum stress in cerebellar development. J Neurosci 2004; 24:1486-96. [PMID: 14960622 PMCID: PMC6730325 DOI: 10.1523/jneurosci.4029-03.2004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The endoplasmic reticulum (ER) stress response contributes to neuronal survival in ischemia and neurodegenerative processes. ORP150 (oxygen-regulated protein 150)/HSP12A (heat shock protein 12A), a novel stress protein located in the ER, was markedly induced in Purkinje cells maximally at 4-8 d after birth, a developmental period corresponding to their vulnerability to cell death. Both terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end-labeling analysis and immunostaining using anti-activated caspase-3 antibody revealed that transgenic mice with targeted neuronal overexpression of ORP150 (Tg ORP150) displayed diminished cell death in the Purkinje cell layer and increased numbers of Purkinje cells up to 40 d after birth (p < 0.01), compared with those observed in heterozygous ORP150/HSP12A-deficient (ORP150+/-) mice and wild-type littermates (ORP150+/+). Cultured Purkinje cells from Tg ORP150 mice displayed resistance to both hypoxia- and AMPA-induced stress. Behavioral analysis, using rotor rod tasks, indicated impairment of cerebellar function in Tg ORP150 animals, consistent with the concept that enhanced survival of Purkinje cells results in dysfunction. These data suggest that ER chaperones have a pivotal role in Purkinje cell survival and death and thus may highlight the importance of ER stress in neuronal development.
Collapse
Affiliation(s)
- Yasuko Kitao
- Department of Neuroanatomy, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8640, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Chen Z, Ljunggren HG, Zhu SW, Winblad B, Zhu J. Reduced susceptibility to kainic acid-induced excitoxicity in T-cell deficient CD4/CD8(-/-) and middle-aged C57BL/6 mice. J Neuroimmunol 2004; 146:33-8. [PMID: 14698844 DOI: 10.1016/j.jneuroim.2003.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Kainic acid (KA)-induced hippocampal injury is a good model for studying human neurodegenerative diseases. To investigate the roles of immune cells and age related changes in neurodegeneration, we used this model to assess reactions in young and middle-aged wild-type and CD4/CD8(-/-) mice by intranasal administration of KA. We found that CD4/CD8-deficiency resulted in a significant reduction of the severity of clinical signs and pathological changes in KA-treated young, but not in KA-treated middle-aged mice. Middle-aged wild-type mice had a similar reaction to KA insult as young and middle-aged CD4/CD8(-/-) mice. CD4/CD8(-/-) mice exhibited decreased locomotor and rearing activities as they approached to middle-aged state, which was not seen in wild-type mice. In addition, CD4/CD8-deficiency and increased age prevented KA-induced increase of both locomotor and rearing activities. The results suggest that a decline of immunological function is associated with aging, and both of them may contribute to the relative resistance to KA-induced neurotoxicity.
Collapse
Affiliation(s)
- Zhiguo Chen
- Division of Experimental Geriatrics, Department of Neurotec, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
36
|
Chen Z, Yu S, Concha HQ, Zhu Y, Mix E, Winblad B, Ljunggren HG, Zhu J. Kainic acid-induced excitotoxic hippocampal neurodegeneration in C57BL/6 mice: B cell and T cell subsets may contribute differently to the pathogenesis. Brain Behav Immun 2004; 18:175-85. [PMID: 14759595 DOI: 10.1016/s0889-1591(03)00117-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2003] [Revised: 06/13/2003] [Accepted: 07/21/2003] [Indexed: 01/06/2023] Open
Abstract
The roles of T cells and B cells in kainic acid (KA)-induced hippocampal lesions were studied in C57BL/6 mice lacking specific T cell populations (CD4, CD8, and CD4/CD8 cells) and B cells [Igh-6(-/-)]. At 48 mg/kg of KA administrated intranasally, KA-induced convulsions were seen in all groups. However, CD4/CD8(-/-) mice exhibited the mildest seizures; the responses of CD8(-/-), Igh-6(-/-) and wild-type mice were intermediate, whereas CD4(-/-) mice displayed much more severe clinical signs and 100% early mortality, indicating that a deficiency of CD4 T cells obviously increased susceptibility to KA-induced brain damage. Histopathological analysis of the mice that survived 7 days after KA administration revealed that CD4/CD8(-/-) mice had the fewest pathologic changes but Igh-6(-/-) mice showed more severe lesions in area CA3 of the hippocampus than CD8(-/-) and wild-type mice. Reactive astrogliosis were prominent in all KA-treated mice. Locomotor activity as assessed by open-field test increased after KA administration in Igh-6(-/-) and wild-type mice only. These results denote the influence of the adaptive immune response on KA-induced hippocampal neurodegeneration and suggest that B cell and T cell subsets may contribute differently to the pathogenesis.
Collapse
Affiliation(s)
- Zhiguo Chen
- Division of Experimental Geriatrics, Department of Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Schauwecker PE, Williams RW, Santos JB. Genetic control of sensitivity to hippocampal cell death induced by kainic acid: A quantitative trait loci analysis. J Comp Neurol 2004; 477:96-107. [PMID: 15281082 DOI: 10.1002/cne.20245] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Host genetic factors are likely to contribute to differences in individual susceptibility to seizure-induced excitotoxic neuronal damage. Similarly, inbred strains of mice differ in their susceptibility to the kainic acid (KA) model of seizure-induced cell death, but the genes responsible for the differences are not known. Here, we define the inheritance patterns of susceptibility to KA-induced neurodegeneration in the hippocampus by assessing 331 back-cross (N2) progeny of two inbred mouse strains, C57BL/6 and FVB/N, previously shown to display resistance and sensitivity to KA-induced cell death, respectively. Results of phenotypic analysis suggest that the difference in susceptibility between these two strains is conferred by a single dominant gene. Therefore, we used an N2 back-cross between the inbred C57BL/6 and FVB/N strains for a genome-wide search for quantitative trait loci (QTLs), which are chromosomal sites containing genes influencing the magnitude of susceptibility. Genome-wide interval mapping in N2 progeny identified a locus on distal chromosome (Chr) 18 with a peak LOD score of 4.9 localized between D18Mit186 and D18Mit4 as having the strongest and most significant effect in this model. QTLs of minor effect were detected on Chr 15 (D15Mit174-D15Mit156) and Chr 4 (D4Mit264-D4Mit91), with peak LOD scores of 3.02 and 2.46, respectively. The three significant QTLs (Chrs 4, 15, 18) together account for nearly 25% of the trait variance for both genders combined. Reduced KA-induced cell death susceptibility was observed in a congenic strain in which the highly susceptible FVB/N strain carried putative resistance alleles from the C57BL/6 strain on Chr 18.
Collapse
Affiliation(s)
- Paula Elyse Schauwecker
- Department of Cell and Neurobiology, University of Southern California, Keck School of Medicine, Los Angeles, California 90089-9112, USA.
| | | | | |
Collapse
|
38
|
Syntichaki P, Tavernarakis N. The biochemistry of neuronal necrosis: rogue biology? Nat Rev Neurosci 2003; 4:672-84. [PMID: 12894242 DOI: 10.1038/nrn1174] [Citation(s) in RCA: 137] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Popi Syntichaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Vassilika Vouton, P.O. Box 1527, Heraklion 71110, Crete, Greece
| | | |
Collapse
|
39
|
Chen Z, Yu S, Bakhiet M, Winblad B, Zhu J. The chemokine receptor CCR5 is not a necessary inflammatory mediator in kainic acid-induced hippocampal injury: evidence for a compensatory effect by increased CCR2 and CCR3. J Neurochem 2003; 86:61-8. [PMID: 12807425 DOI: 10.1046/j.1471-4159.2003.01807.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Chemokines and their receptors have been strongly implicated in the inflammatory process. However, their roles in excitotoxic brain injury are largely unknown. In this study we used C-C chemokine receptor 5 (CCR5) knockout (KO) mice to investigate the role of CCR5 in neurodegeneration induced by intranasal administration of the excitotoxin kainic acid (KA). Although KA treatment resulted in an increased CCR5 mRNA level in the hippocampi of wild-type mice, a CCR5 deficiency in KO mice did not affect either the clinical and pathological changes in vivo or the neuronal susceptibilities to KA insult in vitro. KA treatment stimulated mRNA expression of the monocyte chemoattractant protein-2 (MCP-2) in both the wild-type and KO mice. KA treatment did not affect mRNA levels for the macrophage inflammatory protein-1alpha (MIP-1alpha) or the regulated upon activation normal T cells expressed and secreted protein (RANTES) in either wild-type or CCR5 KO mice. CCR2 mRNA expression was undetectable in the hippocampi of wild-type mice regardless of KA treatment. In contrast, CCR5 KO mice showed CCR2 mRNA expression that was remarkably increased after KA treatment. KA treatment did not affect CCR3 mRNA expression in the wild-type mice, whereas KO mice showed both a higher basal level of CCR3 mRNA expression as well as a strong upregulation following KA treatment. These results indicate that CCR5 is not a necessary inflammatory mediator in KA induced neurodegeneration. The roles of CCR5 in excitotoxic injury in CCR5 deficient mice are compensated by increased CCR2 and CCR3 expression, which share the common MCP-2 ligand with CCR5.
Collapse
MESH Headings
- Animals
- Cell Survival/drug effects
- Cells, Cultured
- Disease Progression
- Excitatory Amino Acid Agonists
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
- Inflammation Mediators/physiology
- Kainic Acid
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurodegenerative Diseases/chemically induced
- Neurodegenerative Diseases/pathology
- Neurodegenerative Diseases/physiopathology
- Neurons/cytology
- Neurons/drug effects
- Neurons/pathology
- RNA, Messenger/metabolism
- Receptors, CCR2
- Receptors, CCR3
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/physiology
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
Collapse
Affiliation(s)
- Zhiguo Chen
- Division of Geriatric Medicine, Department of Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
40
|
Akbar MT, Lundberg AMC, Liu K, Vidyadaran S, Wells KE, Dolatshad H, Wynn S, Wells DJ, Latchman DS, de Belleroche J. The neuroprotective effects of heat shock protein 27 overexpression in transgenic animals against kainate-induced seizures and hippocampal cell death. J Biol Chem 2003; 278:19956-65. [PMID: 12639970 DOI: 10.1074/jbc.m207073200] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The 27-kDa heat shock protein (HSP27) has a potent ability to increase cell survival in response to a wide range of cellular challenges. In order to investigate the mode of action of HSP27 in vivo, we have developed transgenic lines, which express human HSP27 at high levels throughout the brain, spinal cord, and other tissues. In view of the particular property of HSP27 compared with other HSPs to protect neurons against apoptosis, we have tested these transgenic lines in a well established in vivo model of neurotoxicity produced by kainic acid, where apoptotic cell death occurs. Our results demonstrate for the first time the marked protective effects of HSP27 overexpression in vivo, which significantly reduces kainate-induced seizure severity and mortality rate (>50%) in two independent lines and markedly reduces neuronal cell death in the CA3 region of hippocampus. This reduced seizure severity in HSP27 transgenic animals was associated with a marked attenuation of caspase 3 induction and apoptotic features. These studies clearly demonstrate that HSP27 has a major neuroprotective effect in the central nervous system in keeping with its properties demonstrated in culture and highlight an early stage in the cell death pathway that is affected by HSP27.
Collapse
Affiliation(s)
- Mohammed T Akbar
- Department of Neuromuscular Diseases, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|