349 results on '"Neurotoxins"'
Search Results
2. In-vivo comparison of the neurotoxic potencies of incobotulinumtoxinA, onabotulinumtoxinA, and abobotulinumtoxinA
- Author
-
Marie-Christine Reinert, Angelika Mönnich, Anja Manig, David Liebetanz, and Anna Kutschenko
- Subjects
Male ,0301 basic medicine ,medicine.medical_treatment ,Neurotoxins ,Motor Activity ,Pharmacology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,medicine ,Animals ,Potency ,Motor activity ,Botulinum Toxins, Type A ,Saline ,Paresis ,Dose-Response Relationship, Drug ,Botulinum Neurotoxin Type A ,business.industry ,General Neuroscience ,Hindlimb ,3. Good health ,Mice, Inbred C57BL ,Clinical Practice ,030104 developmental biology ,Anesthesia ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Equivalence ratio - Abstract
Three botulinum neurotoxin type A (BoNT/A) products, incobotulinumtoxinA, onabotulinumtoxinA, and abobotulinumtoxinA, all manufactured by different methods, are employed in clinical practice. Comparing the three BoNT/A products is difficult because their concentrations and volumes differ and the precise dose equivalence ratio is not known. We aimed to compare the neurotoxic potencies by a systematic analysis of injected volume and dose. The potency of BoNT in inducing hind limb paresis was assessed by analyzing the wheel-running performance of mice. To standardize the volume, the effect of an identical dose of incobotulinumtoxinA dissolved in different volumes of saline (15, 10, 5, and 2μl) was studied in four groups of mice (n=13-15). The potencies of the BoNT products were then compared by injecting identical volumes (10μl) containing different doses into both hind leg muscles. Mice injected with incobotulinumtoxinA showed a volume-dependent reduction in wheel-running, with larger volumes inducing more intense paresis. A standardized volume containing the same number of mouse units of the BoNT/A products produced different degrees of paresis. The conversion ratio of incobotulinumtoxinA and onabotulinumtoxinA is estimated to be between 1:0.75 and 1:0.5. OnabotulinumtoxinA displayed a two-fold greater potency than abobotulinumtoxinA. Doses of onabotulinumtoxinA and abobotulinumtoxinA that produce an identical severity of pareses even result in the same duration of pareses. This wheel-running assay allows one to compare the neurotoxic potency of different volumes and doses of the BoNT products in vivo. Our results argue against common clinical practice because incobotulinumtoxinA and onabotulinumtoxinA are not readily interchangeable and a two-fold dose of abobotulinumtoxinA is needed to induce an effect identical to onabotulinumtoxinA. In addition, this emphasizes that the duration of BoNT-induced effect is the same as long as equipotent doses of BoNT are injected.
- Published
- 2016
3. Toxins in Neurobiology: New tools from old molecules
- Author
-
Irina Vetter
- Subjects
0301 basic medicine ,Drug discovery ,General Neuroscience ,Production cost ,Neurotoxins ,Vertebrate Animals ,Prey capture ,Biology ,03 medical and health sciences ,Multicellular organism ,030104 developmental biology ,0302 clinical medicine ,Neuropharmacology ,Neurobiology ,Molecular Probes ,Drug Discovery ,Humans ,Evolutionary selection ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Molecules with neurotoxic properties are nearly ubiquitous in nature and are produced by some simple unicellular organisms including algae and bacteria, as well as complex multicellular organisms ranging from plants to invertebrate and vertebrate animals. While the biological roles of many neurotoxins are unknown, and some are simply metabolic waste products, it is perhaps not surprising that most toxins likely provide important survival advantages given the often substantial metabolic production cost to toxic organisms. Targeting mechanisms of neuronal excitability, whether via channel- or receptor-specific effects, or through modulation of fundamental processes such as neurotransmitter release, has immediate and profound effects on many organisms from diverse phyla. Ingested, injected, inhaled, or topical neurotoxins thus cause a range of detrimental consequences in poisoned or envenomated victims, from eliciting pain to complete neuromuscular paralysis. Such effects facilitate prey capture and provide protection from predators or pests, contributing to evolutionary selection of neurotoxic properties over millennia.
- Published
- 2018
4. Effects of tityustoxin on cerebral inflammatory response in young rats
- Author
-
Ricardo Nascimento Araújo, Patrícia Alves Maia Guidine, Rosa Maria Esteves Arantes, Iva Tereza Van Fraga, Tasso Moraes-Santos, André R. Massensini, Marcelo Limborço-Filho, Juliana Carvalho-Tavares, Márcio Flávio Dutra Moraes, Onésia Cristina Oliveira Lima, and Norinne Lacerda-Queiroz
- Subjects
Male ,Scorpion envenoming ,Tityus serrulatus ,Brain inflammation ,Neuroscience(all) ,Inflammatory response ,Neurotoxins ,Scorpion Venoms ,Venom ,Leukocyte Rolling ,Scorpion stings ,Pharmacology ,Microcirculation ,Tityustoxin ,chemistry.chemical_compound ,TsTX ,Leukocyte Trafficking ,Cell Adhesion ,Leukocytes ,Animals ,Medicine ,Rats, Wistar ,Scorpion Stings ,biology ,Tumor Necrosis Factor-alpha ,business.industry ,General Neuroscience ,Leukocyte trafficking ,Brain ,biology.organism_classification ,medicine.disease ,chemistry ,Microvessels ,Immunology ,Encephalitis ,business - Abstract
Accidents caused by scorpion stings, mainly affecting children, are considered an important cause of morbidity and mortality in tropical countries. Clinical studies demonstrate the relevant role of systemic inflammatory events in scorpion envenoming. However, remains poorly understood whether the major lethal component in Tityus serrulatus venom, tityustoxin (TsTX), is able to induce inflammatory responses in the cerebral microcirculation. In this study, we systematically examined leukocyte recruitment into the CNS in response to TsTX injection. Accordingly, developing rats were subjected to a subcutaneous (s.c.) injection of TsTX (0.75 mg/kg), and leukocyte recruitment (i.e., 4, 8 and 12 h after injection) and TNF-α levels were evaluated. Rats injected with TsTX presented a significant increase in leukocyte rolling and adhesion and higher levels of TNF-α at all time points studied, compared to the control group. Altogether, this work demonstrates the triggering of neuroimmunological mechanisms induced by TsTX injection in young rats.
- Published
- 2015
5. Toxins as tools: Fingerprinting neuronal pharmacology
- Author
-
Jennifer R. Deuis, Mathilde R. Israel, Bryan Tay, and Michael M. Morgan
- Subjects
0301 basic medicine ,Sensory Receptor Cells ,Cell ,Neurotoxins ,Computational biology ,Ion Channels ,03 medical and health sciences ,0302 clinical medicine ,Neuropharmacology ,medicine ,Animals ,Humans ,Receptor ,Ion channel ,Neurons ,Voltage-gated ion channel ,Chemistry ,General Neuroscience ,Electrophysiology ,Multicellular organism ,030104 developmental biology ,medicine.anatomical_structure ,Behavior Rating Scale ,Models, Animal ,Ligand-gated ion channel ,Ion Channel Gating ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Toxins have been used as tools for decades to study the structure and function of neuronal ion channels and receptors. The biological origin of these toxins varies from single cell organisms, including bacteria and algae, to complex multicellular organisms, including a wide variety of plants and venomous animals. Toxins are a structurally and functionally diverse group of compounds that often modulate neuronal function by interacting with an ion channel or receptor. Many of these toxins display high affinity and exquisite selectivity, making them valuable tools to probe the structure and function of neuronal ion channels and receptors. This review article provides an overview of the experimental techniques used to assess the effects that toxins have on neuronal function, as well as discussion on toxins that have been used as tools, with a focus on toxins that target voltage-gated and ligand-gated ion channels.
- Published
- 2017
6. Labelled animal toxins as selective molecular markers of ion channels: Applications in neurobiology and beyond
- Author
-
Alexey I. Kuzmenkov and Alexander A. Vassilevski
- Subjects
0301 basic medicine ,Halogenation ,Sensory Receptor Cells ,Drug discovery ,General Neuroscience ,Neurotoxins ,Biology ,Diagnostic tools ,Ion Channels ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Neurobiology ,Molecular targets ,Animals ,Neuroscience ,030217 neurology & neurosurgery ,Ion channel ,Biomarkers ,Fluorescent Dyes - Abstract
Animal toxins are traditional and indispensible molecular tools that find application in different fields of biochemistry, neurobiology and pharmacology. These compounds possess several outstanding properties such as high affinity and selectivity with respect to particular molecular targets, most importantly ion channels and neuroreceptors, and stability. In addition to using toxins per se, a wide variety of labelled modifications have been obtained including radioactive and fluorescent derivatives. Here, we discuss the major types of labelled toxins, methods of their production and principal possibilities of application ranging from receptor localization and visualization to development of screening systems and diagnostic tools, and drug discovery.
- Published
- 2017
7. TGF-β induced by interleukin-34-stimulated microglia regulates microglial proliferation and attenuates oligomeric amyloid β neurotoxicity
- Author
-
Shijie Jin, Di Ma, Yukiko Doi, Yoshifumi Sonobe, Endong Li, Akio Suzumura, Tetsuya Mizuno, and Hideyuki Takeuchi
- Subjects
Neurotoxins ,Neuroprotection ,Mice ,Transforming Growth Factor beta ,medicine ,Insulin-degrading enzyme ,Animals ,Cells, Cultured ,Cell Proliferation ,Amyloid beta-Peptides ,Microglia ,biology ,Chemistry ,Kinase ,Interleukins ,General Neuroscience ,Neurotoxicity ,Interleukin ,Transforming growth factor beta ,medicine.disease ,Cell biology ,medicine.anatomical_structure ,Interleukin 34 ,biology.protein ,Neuroscience - Abstract
Microglia play critical roles in the pathogenesis of Alzheimer's disease (AD). We have previously shown that interleukin-34 (IL-34) enhances microglial proliferation and induces microglial neuroprotective properties against oligomeric amyloid β (oAβ) toxicity by producing insulin degrading enzyme, an Aβ degrading enzyme, and anti-oxidant enzyme heme oxygenase-1. In this study, we found that IL-34 dose-dependently induces TGF-β in microglia, and that TGF-β attenuates oAβ neurotoxicity in neuron microglial co-cultures. The TGF-β 1 receptor kinase inhibitor SD208 enhances microglial proliferation by IL-34 and suppresses the neuroprotective effect of IL-34-treated microglia. These findings suggest that TGF-β produced by IL-34-treated microglia is a negative regulator of microglial proliferation and enhances the neuroprotective property of microglia.
- Published
- 2012
8. Effects of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on retinal dopaminergic system in mice
- Author
-
Syed F. Ali, Merle G. Paule, Bonnie L. Robinson, W. Ryan Hamilton, and William J. Trickler
- Subjects
Male ,Dopamine ,Dopamine Agents ,Neurotoxins ,Pharmacology ,Catechol O-Methyltransferase ,Retina ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,biology ,Chemistry ,General Neuroscience ,MPTP ,Dopaminergic ,Homovanillic acid ,Neurotoxicity ,Homovanillic Acid ,Meth ,Methamphetamine ,medicine.disease ,nervous system diseases ,Mice, Inbred C57BL ,nervous system ,1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine ,biology.protein ,Monoamine oxidase A ,medicine.drug - Abstract
The neurotoxins methamphetamine (METH) and MPTP are well-known for their effects on the nigrostriatal dopaminergic system and use in modeling neurodegenerative disorders such as Parkinson's disease. It is not well-known though, how METH or MPTP affects the visual system and specifically the retinal dopaminergic system. This study was designed to examine acute effects of multiple doses of METH and MPTP on the retinal dopaminergic system. Mice were exposed to either low- (LD) 10 mg/kg total dose or high-dose (HD) 30 mg/kg total dose, of METH or MPTP and the retinal catecholaminergic system was analyzed by HPLC. METH produced no significant changes in dopamine (DA), its metabolites 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) or DA usage in the retina. LD-MPTP produced no change in DA level, but significantly decreased DOPAC and HVA. LD-MPTP also significantly decreased DA usage as measured by the DOPAC/DA and HVA/DA ratios. HD-MPTP significantly decreased DA, DOPAC and HVA, but did not affect DA usage. Taken together these results suggest that inhibition of the DA metabolizing enzymes monoamine oxidase A (MAO) or catechol-O-methyl transferase (COMT) may take place at lower doses of MPTP treatment; conversely, higher doses of MPTP may cause decreases in DA, DOPAC and HVA through another mechanism.
- Published
- 2012
9. Superior colliculus lesions impair threat responsiveness in infant capuchin monkeys
- Author
-
Marilia Barros, Hisao Nishijo, Maria Clotilde H. Tavares, Rafael S. Maior, Taketoshi Ono, Danilo Simonini Teixeira, Carlos Tomaz, and Etsuro Hori
- Subjects
Long lasting ,Superior Colliculi ,Neurotoxins ,Klüver–Bucy syndrome ,Amygdala ,Developmental psychology ,Conflict, Psychological ,chemistry.chemical_compound ,Reward ,Escape Reaction ,Reaction Time ,medicine ,Biological neural network ,Animals ,Cebus ,Humans ,Ibotenic Acid ,Facial expression ,General Neuroscience ,Superior colliculus ,Emotional stimuli ,Snakes ,Fear ,Feeding Behavior ,medicine.disease ,Disease Models, Animal ,Inhibition, Psychological ,Instillation, Drug ,medicine.anatomical_structure ,Pattern Recognition, Visual ,chemistry ,Kluver-Bucy Syndrome ,Psychology ,Neuroscience ,psychological phenomena and processes ,Ibotenic acid - Abstract
The ability to react fast and efficiently in threatening situations is paramount for the survival of organisms and has been decisive in our evolutionary history. Defense mechanisms in primates rely on the fast recognition of potential predators and facial expressions of conspecifics. The neural circuitry responsible for the detection of threat is generally thought to be centered on the amygdala. Although it is a pivotal structure in the processing of emotional stimuli, the amygdala does not seem necessary for the early stages of this process. Here we show that bilateral neurotoxic lesions of the superior colliculus in infant capuchins monkeys impaired the recognition of a rubber-snake in a threat-reward conflict task. Lesioned monkeys were uninhibited by a snake in a food-reward retrieval task. Lack of inhibition in the task was observed over the course of 15 weeks. The long lasting recognition impairment of a natural predator observed here is similar to the tameness aspects of Kluver-Bucy syndrome, indicating an important role of this structure in threat recognition.
- Published
- 2011
10. Rotenone induced neurotoxicity in rat brain areas: A histopathological study
- Author
-
Sarika Singh, Ishan K. Patro, Chandishwar Nath, Sharad Sharma, Ramesh Mathur, and Supriya Swarnkar
- Subjects
Male ,Dyskinesia, Drug-Induced ,Insecticides ,Pathology ,medicine.medical_specialty ,Neurotoxins ,H&E stain ,Substantia nigra ,Striatum ,Biology ,Rats, Sprague-Dawley ,Cresyl violet ,chemistry.chemical_compound ,Rotenone ,medicine ,Animals ,Neurons ,Dose-Response Relationship, Drug ,General Neuroscience ,Neurotoxicity ,Brain ,medicine.disease ,Corpus Striatum ,Rats ,Staining ,Motor coordination ,Substantia Nigra ,chemistry ,Nerve Degeneration ,sense organs - Abstract
Rotenone a pesticide is known to induce neurotoxicity. In earlier study we correlated rotenone induced biochemical changes and cerebral damage in brain areas with neuromuscular coordination in rats. The present study involves investigation of rotenone induced histopathological changes in the brain areas, viz. striatum (STR) and substantia nigra (SN) using HE (hematoxylin and eosin) and CV (Cresyl Violet) staining after 1, 7, and 14 day of unilateral intranigral administration of rotenone (3, 6 and 12 μg/5 μl) in adult male SD rats. Significant morphological changes in cell area or shape were shown by HE staining. The neuronal degeneration was shown by distorted neuronal cells, shrinkage of nuclei, dark staining in the regions of rotenone treated animals by CV staining. Rota rod test demonstrated significant impairment in motor coordination after 14 days of treatment along with decreased GSH and increased MDA in STR and mid brain (MB). The study inferred rotenone causes neuronal damage which is evident by histopathological changes, impaired neuromuscular coordination and biochemical changes. The pattern of histopathological alterations corresponds with behavioral and biochemical manifestations.
- Published
- 2011
11. Neuroprotective effects of tenuigenin in a SH-SY5Y cell model with 6-OHDA-induced injury
- Author
-
Xuan Wang, Zhi-Gang Liang, Li Lu, Fang Shi, Zhanjun Zhang, Xiaomin Wang, and Yong Wang
- Subjects
medicine.medical_specialty ,Programmed cell death ,SH-SY5Y ,Neurotoxins ,Pharmacology ,Neuroprotection ,Antioxidants ,Superoxide dismutase ,chemistry.chemical_compound ,Cell Line, Tumor ,Internal medicine ,medicine ,Humans ,Viability assay ,Oxidopamine ,Neurons ,biology ,Tyrosine hydroxylase ,General Neuroscience ,Parkinson Disease ,Glutathione ,Neural stem cell ,Neuroprotective Agents ,Endocrinology ,chemistry ,biology.protein ,Drugs, Chinese Herbal - Abstract
Tenuigenin, an active component of Polygala tenuifolia root extracts, has been shown to provide antioxidative and anti-aging effects in Alzheimer's disease, as well as to promote proliferation and differentiation of neural progenitor cells. However, the effects of tenuigenin on Parkinson's disease remain unclear. In the present study, SH-SY5Y cells were utilized to determine the effects of tenuigenin on 6-hydroxydopamine (6-OHDA)-induced injury. Results showed that 1.0 × 10⁻¹-10 μM tenuigenin significantly promoted cell viability and reduced cell death. In addition, tenuigenin protected mitochondrial membrane potential (MMP) against 6-OHDA damage and significantly increased glutathione and superoxide dismutase expression. At the mRNA level, tenuigenin resulted in down-regulation of caspase-3, but up-regulation of tyrosine hydroxylase expression in 6-OHDA damaged cells. These results suggested that tenuigenin provides neuroprotection to dopaminergic neurons from 6-OHDA-induced damage. The neuroprotective mechanisms might involve antioxidative effects, maintenance of mitochondrial function, and regulation of caspase-3 and tyrosine hydroxylase expression and activity. Tenuigenin could provide a novel antioxidative strategy for Parkinson's disease.
- Published
- 2011
12. Brain-derived neurotrophic factor (BDNF) ameliorates the suppression of thyroid hormone-induced granule cell neurite extension by hexabromocyclododecane (HBCD)
- Author
-
Kingsley Ibhazehiebo, Noriyuki Koibuchi, Noriaki Shimokawa, Toshiharu Iwasaki, and Ming Xu
- Subjects
Thyroid Hormones ,Cerebellum ,medicine.medical_specialty ,Neurite ,Cellular differentiation ,Neurotoxins ,Central nervous system ,Antithyroid Agents ,Neurotrophic factors ,Internal medicine ,Neurites ,medicine ,Animals ,Rats, Wistar ,Receptor ,Cells, Cultured ,Brain-derived neurotrophic factor ,Chemistry ,Brain-Derived Neurotrophic Factor ,General Neuroscience ,Cell Differentiation ,Granule cell ,Hydrocarbons, Brominated ,Rats ,Neuroprotective Agents ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn - Abstract
Thyroid hormone (TH) plays an essential role in growth and differentiation of the central nervous system. Deficiency of TH during perinatal period results in abnormal brain development known as cretinism in human. We recently reported that an environmental chemical 1,2,5,6,9,10-α-hexabromocyclododecane (HBCD) suppressed TH receptor (TR)-mediated transcription. To examine the effect of HBCD on cerebellar granule cells, we used purified rat cerebellar granule cells in reaggregate culture. Low dose HBCD (10(-10)M) significantly suppressed TH-induced neurite extension of granule cell aggregate. To clarify further the mechanisms of such suppression, we added brain-derived neurotrophic factor (BDNF) into culture medium, since BDNF plays a critical role in promoting granule cell development and is regulated by TH. BDNF completely rescued HBCD-induced suppression of granule cell neurite extension in the presence of T3. These results indicate that HBCD may disrupt TH-mediated brain development at least in part due to a disruption of the T3 stimulated increase in BDNF and BDNF may possess ability to ameliorate the effect of HBCD in granule cells.
- Published
- 2011
13. Implication of the c-Jun-NH2-terminal kinase pathway in the neuroprotective effect of puerarin against 1-methyl-4-phenylpyridinium (MPP+)-induced apoptosis in PC-12 cells
- Author
-
Xiaoming Li, Yingcai Niu, Li Zhou, Miaoxian Dong, Ying-Bo Zhang, Jicheng Liu, and Gang Wang
- Subjects
medicine.medical_specialty ,Programmed cell death ,Cell Survival ,Neurotoxins ,Apoptosis ,Pharmacology ,PC12 Cells ,Neuroprotection ,Drug Administration Schedule ,chemistry.chemical_compound ,Puerarin ,Annexin ,Internal medicine ,medicine ,Animals ,Drug Interactions ,MTT assay ,Annexin A5 ,Enzyme Inhibitors ,Dose-Response Relationship, Drug ,L-Lactate Dehydrogenase ,biology ,Caspase 3 ,General Neuroscience ,Cytochrome c ,JNK Mitogen-Activated Protein Kinases ,Neurotoxicity ,medicine.disease ,Isoflavones ,Rats ,Neuroprotective Agents ,Endocrinology ,nervous system ,chemistry ,1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine ,biology.protein ,Signal Transduction - Abstract
Apoptosis is a widely accepted component of the pathogenesis of Parkinson's disease (PD), a debilitating neurodegenerative disorder characterized by loss of dopaminergic neurons in the substantia nigra. In this study, we investigated the neuroprotective effects of puerarin and possible mechanisms by which puerarin acts against MPP(+)-induced toxicity in rat pheochromocytoma PC12 cells. PC12 cells exposed to MPP(+) (500 μM) significantly decreased the viability of PC12 cells when examined by MTT assay, DNA ELISA assay, and Annexin V assays, which was prevented by puerarin in a dose-dependent manner. PC12 cells exposed to MPP(+) (500 μM) elicited phosphorylation of MKK7, c-Jun-NH(2)-terminal kinase (JNK), and c-Jun which followed by the increase in cytochrome c levels, and which was prevented by puerarin. Moreover, puerarin inhibited the activation of caspase-9 and caspase-3 in MPP(+)-exposed PC12 cells. Whereas, the neuroprotective effect of puerarin against MPP(+) insults can be blocked by SP600125 (inhibitor of JNK). Taken together, these results suggest that puerarin protected PC12 cells against MPP(+)-induced neurotoxicity through the inhibition of the JNK signaling pathways. Therefore, puerarin has the possible beneficial effects in PD by attenuating MPP(+)-induced toxicity.
- Published
- 2011
14. Zonisamide reduces cell death in SH-SY5Y cells via an anti-apoptotic effect and by upregulating MnSOD
- Author
-
Nobutaka Hattori, Shigeto Sato, Yutaka Machida, Shinji Saiki, and Sumihiro Kawajiri
- Subjects
medicine.medical_specialty ,Levodopa ,Programmed cell death ,SH-SY5Y ,Dopamine ,Neurotoxins ,Zonisamide ,Apoptosis ,Pharmacology ,Antioxidants ,Superoxide dismutase ,Neuroblastoma ,Internal medicine ,In Situ Nick-End Labeling ,medicine ,Humans ,Drug Interactions ,Viability assay ,Enzyme Inhibitors ,Dose-Response Relationship, Drug ,biology ,Superoxide Dismutase ,business.industry ,General Neuroscience ,Neurotoxicity ,Cell Differentiation ,Isoxazoles ,Staurosporine ,medicine.disease ,Up-Regulation ,Endocrinology ,1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine ,Caspases ,biology.protein ,business ,medicine.drug - Abstract
Zonisamide, originally known as an antiepileptic drug, has been approved in Japan as adjunctive therapy with levodopa for the treatment of Parkinson's disease (PD). Although zonisamide reduces neurotoxicity, the precise mechanism of this action is not known. Here, we show that zonisamide increases cell viability in SH-SY5Y cells via an anti-apoptotic effect and by upregulating levels of manganese superoxide dismutase (MnSOD). These results would give us novel evidences of PD treatment.
- Published
- 2010
15. Neuroprotective effects of pyruvate following NMDA-mediated excitotoxic insults in hippocampal slices
- Author
-
Yukitoshi Izumi and Charles F. Zorumski
- Subjects
Aging ,N-Methylaspartate ,Neurotoxins ,Excitotoxicity ,Hippocampus ,In Vitro Techniques ,Hippocampal formation ,Biology ,Nitric Oxide ,medicine.disease_cause ,Receptors, N-Methyl-D-Aspartate ,Neuroprotection ,Article ,Nitric oxide ,chemistry.chemical_compound ,Adenosine Triphosphate ,Pyruvic Acid ,medicine ,Animals ,Glycolysis ,Enzyme Inhibitors ,Receptor ,CA1 Region, Hippocampal ,General Neuroscience ,Glyceraldehyde-3-Phosphate Dehydrogenases ,Rats ,nervous system ,chemistry ,Brain Injuries ,Nerve Degeneration ,Synapses ,NMDA receptor ,Excitatory Amino Acid Antagonists ,Neuroscience - Abstract
The activation of N-methyl-D-aspartate (NMDA) receptors and subsequent release of nitric oxide (NO) are likely contributors to the delayed neuronal damage that accompanies ischemia and other neurodegenerative conditions. NMDA receptor antagonists and inhibitors of NO synthesis, however, are of limited benefit when administered following excitotoxic events, suggesting the importance of determining downstream events that result in neuronal degeneration. Inhibition of glyceraldehyde-3-phosphate-dehydrogenase (GAPDH), a key glycolytic enzyme, which may result in glycolytic impairment, is one of the biological targets of NO. This suggests that alternative energy substrates may prevent neuronal damage. Using rat hippocampal slices from juvenile rats, we examined the role of glycolytic impairment in NMDA mediated excitotoxicity and whether pyruvate, an end product of glycolysis, prevents the excitotoxic neuronal injury. We observed that administration of NMDA acutely depresses ATP levels and result in a slowly developing inhibition of GAPDH. Unlike NMDA receptor antagonists or NO inhibitors, exogenously applied pyruvate is effective in restoring ATP levels and preventing delayed neuronal degeneration and synaptic deterioration when administered in the period following NMDA receptor activation. This raises the possibility that treatment with agents that maintain cellular energy function can prevent delayed excitotoxicity.
- Published
- 2010
16. Granulocyte-colony stimulating factor is involved in low-dose LPS-induced neuroprotection
- Author
-
Qin-Ying Li, Zheng-Xi Li, Chuan-Zhen Lu, Bao-Guo Xiao, and Jian Qiao
- Subjects
Lipopolysaccharides ,N-Methylaspartate ,Time Factors ,Lipopolysaccharide ,Cell Survival ,Neurotoxins ,Central nervous system ,Pharmacology ,Hippocampal formation ,Biology ,Hippocampus ,Neuroprotection ,chemistry.chemical_compound ,In vivo ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,RNA, Messenger ,Cells, Cultured ,Neurons ,Messenger RNA ,Dose-Response Relationship, Drug ,General Neuroscience ,Rats ,Granulocyte colony-stimulating factor ,Toll-Like Receptor 4 ,Neuroprotective Agents ,medicine.anatomical_structure ,chemistry ,Immunology ,Neuron - Abstract
Granulocyte-colony stimulating factor (G-CSF) has recently been noted for neuroprotective function. Evidence has been given to indicate that G-CSF is naturally expressed in neurons and directly activates anti-apoptosis pathways. Finding out the agents inducing G-CSF production is of value for understanding the neuroprotection network in central nervous system. It is known that lipopolysaccharide (LPS) can induce macrophages to produce G-CSF. Here we demonstrate that hippocampal neurons exhibited the expression of toll-like receptor-4, and prove that low-dose LPS treatment increased the expression and production of G-CSF mRNA and protein in cultured neurons. We further indicate that the neutralization of G-CSF with corresponding anti-G-CSF antibodies abolished the neuroprotective effect of LPS pretreatment in N-methyl- d -aspartic acid-induced neuronal injury by MTT/CCK-8 assays and LDH release. Thus our results reveal that G-CSF may be involved in LPS-mediated neuroprotection in vivo.
- Published
- 2009
17. Nanoparticle–chelator conjugates as inhibitors of amyloid-β aggregation and neurotoxicity: A novel therapeutic approach for Alzheimer disease
- Author
-
George Perry, Gang Liu, Wataru Kudo, Mark A. Smith, and Ping Men
- Subjects
Amyloid ,Pyridones ,Iron ,Neurotoxins ,Plaque, Amyloid ,Pharmacology ,Iron Chelating Agents ,medicine.disease_cause ,Article ,Alzheimer Disease ,medicine ,Humans ,Chelation ,Cells, Cultured ,Neurons ,Amyloid beta-Peptides ,Chemistry ,General Neuroscience ,Neurodegeneration ,Neurotoxicity ,Brain ,medicine.disease ,Iron Metabolism Disorders ,Nerve Degeneration ,Toxicity ,Drug delivery ,Nanoparticles ,Alzheimer's disease ,Neuroscience ,Oxidative stress - Abstract
Oxidative stress and amyloid-beta are considered major etiological and pathological factors in the initiation and promotion of neurodegeneration in Alzheimer disease (AD). Insomuch as causes of such oxidative stress, transition metals, such as iron and copper, which are found in high concentrations in the brains of AD patients and accumulate specifically in the pathological lesions, are viewed as key contributors to the altered redox state. Likewise, the aggregation and toxicity of amyloid-beta is dependent upon transition metals. As such, chelating agents that selectively bind to and remove and/or "redox silence" transition metals have long been considered as attractive therapies for AD. However, the blood-brain barrier and neurotoxicity of many traditional metal chelators has limited their utility in AD or other neurodegenerative disorders. To circumvent this, we previously suggested that nanoparticles conjugated to iron chelators may have the potential to deliver chelators into the brain and overcome such issues as chelator bioavailability and toxic side-effects. In this study, we synthesized a prototype nanoparticle-chelator conjugate (Nano-N2PY) and demonstrated its ability to protect human cortical neurons from amyloid-beta-associated oxidative toxicity. Furthermore, Nano-N2PY nanoparticle-chelator conjugates effectively inhibited amyloid-beta aggregate formation. Overall, this study indicates that Nano-N2PY, or other nanoparticles conjugated to metal chelators, may provide a novel therapeutic strategy for AD and other neurodegenerative diseases associated with excess transition metals.
- Published
- 2009
18. Ketogenic diet attenuates kainic acid-induced hippocampal cell death by decreasing AMPK/ACC pathway activity and HSP70
- Author
-
Wan Sung Choi, Hyun Joon Kim, Byeong Tak Jeon, Gyeong Jae Cho, Sang Soo Kang, Dong Hoon Lee, Gu Seob Roh, and Kyu Hong Kim
- Subjects
Male ,medicine.medical_specialty ,Kainic acid ,Programmed cell death ,Normal diet ,Blotting, Western ,Neurotoxins ,Gene Expression ,Apoptosis ,AMP-Activated Protein Kinases ,Biology ,Hippocampus ,Mice ,chemistry.chemical_compound ,Seizures ,Internal medicine ,medicine ,Animals ,HSP70 Heat-Shock Proteins ,Phosphorylation ,Protein kinase A ,Analysis of Variance ,Mice, Inbred ICR ,Kainic Acid ,General Neuroscience ,Body Weight ,AMPK ,Hsp70 ,Endocrinology ,chemistry ,Signal transduction ,Diet, Ketogenic ,Acetyl-CoA Carboxylase - Abstract
The ketogenic diet (KD) prevents kainic acid (KA)-induced hippocampal cell death. There are reports that AMP-activated protein kinase (AMPK) activation regulates the intracellular signaling pathways involved in cellular survival or apoptotic cell death. In this study, we investigated the effect of the KD consumption on the expression of signaling pathway proteins AMPK and ACC, and heat shock protein (HSP) 70 in mouse hippocampus after KA treatment. Mice were fed the KD for 6 weeks and then sacrificed 48 h after KA (30 mg/kg) injection. The marked cell death found commonly in normal diet (ND)-fed mice treated with KA was not observed in the KD-fed KA-treated mice. Western blot analysis revealed that phosphorylation of AMPK and ACC was increased after KA treatment. However, phosphorylation of these proteins was reduced in those animals that received the KD. In addition, increased expression of HSP70 in the hippocampus of KA-treated mice was decreased in animals receiving the KD. These results indicate that the KD promotes neuroprotective effects through suppression of the AMPK cascade and that HSP70 is involved in neuronal cell death or oxidative stress.
- Published
- 2009
19. Demyelination of the hippocampus is prominent in the cuprizone model
- Author
-
Paraskevi N. Koutsoudaki, Herbert Hildebrandt, Martin Stangel, Thomas Skripuletz, Corinna Trebst, Darius Moharregh-Khiabani, and Viktoria Gudi
- Subjects
Male ,Neurotoxins ,Hippocampus ,Nerve Tissue Proteins ,Biology ,Grey matter ,Hippocampal formation ,Nerve Fibers, Myelinated ,White matter ,Cuprizone ,Mice ,Myelin ,medicine ,Animals ,Gliosis ,Chelating Agents ,Neocortex ,General Neuroscience ,medicine.disease ,Immunohistochemistry ,Astrogliosis ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Astrocytes ,Microglia ,medicine.symptom ,Wallerian Degeneration ,Neuroscience ,Biomarkers ,Demyelinating Diseases - Abstract
In multiple sclerosis demyelination not only affects the white matter, but also the grey matter of the brain. We have previously reported that in the murine cuprizone model for demyelination lesions occur in addition to the corpus callosum also in the neocortex and hippocampus. In the current study, we provide a detailed characterization of hippocampal demyelination in the cuprizone model. Male C57BL/6 mice were challenged with 0.2% cuprizone for 6 weeks. Defined structures within the hippocampus were investigated at week 0 (control), 3, 4, 4.5, 5, 5.5, and 6. Demyelination affected all hippocampal structures analyzed and was complete after 6 weeks of cuprizone treatment. Between the distinct hippocampal structures the temporal pattern of demyelination varied considerably. Furthermore, infiltration of activated microglia as well as astrogliosis was detected. In summary, cuprizone feeding provides a useful model for studying demyelination processes in the mouse hippocampus.
- Published
- 2009
20. Impaired calcium homeostasis in aged hippocampal neurons
- Author
-
Christian Behl, Parvana Hajieva, Christoph R.W. Kuhlmann, and Heiko J. Luhmann
- Subjects
Aging ,medicine.medical_specialty ,Neurotoxins ,Glutamic Acid ,chemistry.chemical_element ,Hippocampus ,Biology ,Calcium ,Calcium in biology ,Rats, Sprague-Dawley ,Internal medicine ,medicine ,Animals ,Calcium Signaling ,Organic Chemicals ,Cells, Cultured ,Cellular Senescence ,Neurons ,Calcium metabolism ,Calpain ,General Neuroscience ,Neurodegeneration ,Glutamate receptor ,Hydrogen Peroxide ,Oxidants ,medicine.disease ,Rats ,Oxidative Stress ,medicine.anatomical_structure ,Endocrinology ,nervous system ,chemistry ,Nerve Degeneration ,biology.protein ,Neuron ,Neuroscience - Abstract
Development of neurodegenerative diseases such as Alzheimer's and Parkinson's disease is strongly age-associated. The impairment of calcium homeostasis is considered to be a key pathological event leading to neuronal dysfunction and cell death. However, the exact impact of aging on calcium homeostasis in neurons remains largely unknown. In the present work we have investigated intracellular calcium levels in cultured primary hippocampal neurons from young (2 months) and aged (24 months) rat brains. Upon stimulation with glutamate or hydrogen peroxide aged neurons in comparison to young neurons demonstrated an increased vulnerability to these disease-related toxins. Measurement of calpain activity using Western blot analysis showed a significant increase in basal activity of calpains in aged neurons. The observed increase of calpain activity was correlated with elevated protein levels of μ-calpain. Ca 2+ -imaging experiments performed on living individual neurons using the dye calcium green demonstrated a twofold increase in intracellular calcium concentration in aged neurons as compared to young neurons. The observed changes of intracellular calcium in aged neurons might play a role in their increased vulnerability to neurodegeneration.
- Published
- 2009
21. Melatonin prevents ischemic brain injury through activation of the mTOR/p70S6 kinase signaling pathway
- Author
-
Phil-Ok Koh
- Subjects
Male ,medicine.medical_specialty ,Programmed cell death ,Cell Survival ,Neurotoxins ,Biology ,Antioxidants ,Functional Laterality ,Rats, Sprague-Dawley ,Melatonin ,Mice ,Glutamates ,Internal medicine ,medicine ,Animals ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Line, Transformed ,Neurons ,Sirolimus ,Analysis of Variance ,Kinase ,Cell growth ,General Neuroscience ,Ribosomal Protein S6 Kinases, 70-kDa ,Infarction, Middle Cerebral Artery ,Rats ,Disease Models, Animal ,Endocrinology ,Gene Expression Regulation ,Phosphorylation ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
We previously reported that melatonin prevents neuronal cell death in ischemic brain injury through the activation of Akt and the inhibition of apoptotic cell death. We investigated whether melatonin inhibits the apoptotic signal through the activation of a mammalian target of rapamycin (mTOR) and p70S6 kinase and its downstream target, S6 phosphorylation. It is known that mTOR is a downstream target of Akt and a central regulator of protein synthesis, cell growth, and cell cycle progression. Adult male rats were treated with melatonin (5mg/kg) or vehicle prior to middle cerebral artery occlusion (MCAO). Brains were collected at 24h after MCAO and infarct volumes were analyzed. We confirmed that melatonin significantly reduces infarct volume and decreases the number of TUNEL-positive cells in the cerebral cortex. Brain injury induced a decrease in phospho-mTOR and phospho-p70S6 kinase. Melatonin prevented the injury-induced decrease in Akt activation and phosphorylation of mTOR and p70S6 kinases, and the subsequent decrease in S6 phosphorylation. Our results suggest that melatonin prevents cell death resulting from ischemic brain injury and that its neuroprotective effects are mediated by preventing the injury-induced decrease of mTOR and p70S6 kinase phosphorylation.
- Published
- 2008
22. Silencing of peroxiredoxin 3 and peroxiredoxin 5 reveals the role of mitochondrial peroxiredoxins in the protection of human neuroblastoma SH-SY5Y cells toward MPP+
- Author
-
Julie Goemaere, Stéphanie De Simoni, and Bernard Knoops
- Subjects
1-Methyl-4-phenylpyridinium ,Peroxiredoxin III ,SH-SY5Y ,Cell Survival ,Neurotoxins ,Down-Regulation ,Apoptosis ,Mitochondrion ,Biology ,medicine.disease_cause ,Neuroblastoma ,Tumor Cells, Cultured ,medicine ,Humans ,Gene silencing ,Gene Silencing ,RNA, Small Interfering ,Neurons ,General Neuroscience ,PRDX5 ,Peroxiredoxins ,Mitochondria ,Cell biology ,PRDX3 ,Substantia Nigra ,Oxidative Stress ,Electron Transport Chain Complex Proteins ,Biochemistry ,Cytoprotection ,Energy Metabolism ,Peroxiredoxin ,Oxidative stress - Abstract
Peroxiredoxins (PRDXs) are a family of peroxidases well conserved throughout evolution. Human PRDX3 and PRDX5, two mitochondrial PRDXs, have been implicated in several pathologies associated with oxidative stress. However, the individual role of PRDX3 and PRDX5 in cellular antioxidant defense has never been well established due to their overlapping peroxidatic activities. We investigated the expression and function of mitochondrial PRDXs in human neuroblastoma SH-SY5Y cells. Our results show that PRDX3 and PRDX5 are expressed constitutively in these neuronal cells. To examine further the function of mitochondrial PRDXs, we silenced the expression of PRDX3 and/or PRDX5 using small hairpin RNAs. Our results show that mitochondrial PRDX-depleted cells are more prone to oxidative damages and apoptosis induced by MPP(+), a complex I inhibitor which provides an experimental paradigm of Parkinson's disease.
- Published
- 2008
23. PLA2 signaling is involved in calpain-mediated degradation of synaptic dihydropyrimidinase-like 3 protein in response to NMDA excitotoxicity
- Author
-
Kristoffer Laser Moraleja, Balu Chakravarthy, and Renata Kowara
- Subjects
Canada ,N-Methylaspartate ,IBMX ,Cytoskeleton organization ,Neurotoxins ,Excitotoxicity ,Nerve Tissue Proteins ,ORGANIZATION ,medicine.disease_cause ,chemistry.chemical_compound ,Cyclic AMP ,Excitatory Amino Acid Agonists ,medicine ,Animals ,Enzyme Inhibitors ,Protein kinase A ,Cells, Cultured ,Cerebral Cortex ,Neurons ,Forskolin ,Cyclooxygenase 2 Inhibitors ,biology ,Calpain ,General Neuroscience ,Role ,Glutamate receptor ,Brain ,INHIBITOR ,toxicity ,DEGRADATION ,Cyclic AMP-Dependent Protein Kinases ,Rats ,Cell biology ,Enzyme Activation ,Phospholipases A2 ,nervous system ,Biochemistry ,chemistry ,Adenylyl Cyclase Inhibitors ,Synapses ,biology.protein ,NMDA receptor ,INHIBITORS ,protein ,Signal Transduction - Abstract
Dihydropyrimidinase-like 3 (DPYSL3) is believed to play a role in neuronal differentiation, axonal outgrowth and neuronal regeneration, as well as cytoskeleton organization. Recently we have shown that glutamate excitotoxicity and oxidative stress result in calpain-dependent cleavage of DPYSL3, and that NOS plays a role in this process [R. Kowara, Q. Chen, M. Milliken, B. Chakravarthy, Calpain-mediated truncation of dihydropyrimidinase-like 3 protein (DPYSL3) in response to NMDA and H2O2 toxicity, J. Neurochem. 95 (2005) 466-474; R. Kowara, K.L. Moraleja, B. Chakravarthy, Involvement of nitric oxide synthase and ROS-mediated activation of L-type voltage-gated Ca(2+) channels in NMDA-induced DPYSL3 degradation, Brain Res. 1119 (2006) 40-49]. The present study investigates the involvement of PLA(2) signaling in NMDA-induced DPYSL3 degradation. Exposure of rat primary cortical neurons (PCN) to PLA(2) and COX-2 inhibitors significantly prevented NMDA-induced DPYSL3 degradation. Since the metabolic product of PLA(2) signaling, PGE(2), which augments toxic effect of NMDA, is known to stimulate cAMP, the effect of adenyl cyclase activator (forskolin plus IBMX) and inhibitor (MDL12,300) on NMDA-induced DPYSL3 degradation was tested. Our data indicate that the activation of adenyl cyclase contributes to NMDA-induced DPYSL3 degradation. Furthermore, cAMP-dependent protein kinase (PKA) inhibitor PKI (14-22) provided additional evidence of PKA involvement in NMDA-induced DPYSL3 degradation. In summary, the obtained data show the contribution of PLA(2) signaling to NMDA-induced calpain activation and subsequent degradation of synaptic protein DPYSL3.
- Published
- 2008
24. Increased expression of the lysosomal protease cathepsin S in hippocampal microglia following kainate-induced seizures
- Author
-
Yasuki Ishizaki, Isao Ishii, Toshiyuki Himi, Noriyuki Akahoshi, and Yoshiya L. Murashima
- Subjects
Male ,Nicotine ,Kainic acid ,Neurotoxins ,Kainate receptor ,Hippocampal formation ,Biology ,Hippocampus ,Gene Expression Regulation, Enzymologic ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,Gliosis ,Nicotinic Agonists ,Coloring Agents ,Oligonucleotide Array Sequence Analysis ,Cathepsin S ,Cathepsin ,Epilepsy ,Kainic Acid ,Pyramidal Cells ,General Neuroscience ,Neurodegeneration ,medicine.disease ,Cathepsins ,Molecular biology ,Up-Regulation ,Astrogliosis ,Enzyme Activation ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,nervous system ,chemistry ,Astrocytes ,Nerve Degeneration ,Neuroglia ,Microglia ,Lysosomes - Abstract
To examine lesions caused by seizures in the developing brain, seizures were induced by the intraperitoneal injection of kainate and nicotine into juvenile mice. After a week, whole brain sections were examined using histochemistry and the gene expression profiles in the neocortices and hippocampi were analyzed using a DNA microarray. Propidium iodide and Fluoro-Jade C staining revealed that kainate but not nicotine-induced degeneration of the hippocampal pyramidal neurons. Comparative analyses of 12,488 probe sets on the microarray chip revealed the differential expression of 208 and 1243 probe sets in the neocortices and hippocampi of kainate-injected mice, respectively, as well as that of 535 and 436 probe sets in the neocortices and hippocampi of nicotine-injected mice, respectively, the patterns of change were largely drug-specific and region-specific. Among a variety of kainate-modified genes including those representing neurodegeneration and astrogliosis, we identified an increased gene expression of the lysosomal cysteine protease cathepsin S in the hippocampi of kainate-injected mice. Western blot analysis of the hippocampal homogenates revealed that kainate induced a 3.3-fold increase in cathepsin S expression. Immunohistochemistry using cell type-specific markers showed that cathepsin S was induced in microglia, especially those surrounding degenerating pyramidal neurons, but not in neurons themselves or astroglia, in the hippocampal CA1 region of kainate-injected mice. These results indicate that seizures induced by kainate elicit neurodegeneration, astrogliosis, and microglial activation accompanied by the expression of cathepsin S while those induced by nicotine do not.
- Published
- 2007
25. In vitro and ex vivo autoradiography studies on peripheral-type benzodiazepine receptor binding using [11C]AC-5216 in normal and kainic acid-lesioned rats
- Author
-
Ming-Rong Zhang, Kazuhiko Yanamoto, Katsushi Kumata, Akiko Hatori, Maki Okada, and Kazutoshi Suzuki
- Subjects
Male ,Kainic acid ,Neurotoxins ,Antineoplastic Agents ,Striatum ,In Vitro Techniques ,Biology ,Ligands ,Binding, Competitive ,Hippocampus ,Radioligand Assay ,chemistry.chemical_compound ,Excitatory Amino Acid Agonists ,medicine ,Animals ,Carbon Radioisotopes ,Rats, Wistar ,Receptor ,Cerebral Cortex ,Kainic Acid ,Dose-Response Relationship, Drug ,General Neuroscience ,Brain ,Isoquinolines ,Receptors, GABA-A ,Ligand (biochemistry) ,Molecular biology ,Corpus Striatum ,Rats ,Olfactory bulb ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Purines ,Cerebral cortex ,Positron-Emission Tomography ,Autoradiography ,Encephalitis ,Choroid plexus ,Carrier Proteins ,Neuroscience ,Ex vivo - Abstract
AC-5216 was reported as a novel ligand for peripheral-type benzodiazepine receptor (PBR) with a different chemical structure from DAA1106 analogues. This ligand had potent affinity for PBR and selectivity for PBR over other neurotransmitters. We have previously labeled AC-5216 using positron-emitter 11 C. The aim of this study was to evaluate [ 11 C]AC-5216 in a rat brain model with neuroinflammation using an autoradiography (ARG) technique. In vitro ARG of normal rat brain showed that [ 11 C]AC-5216 accumulated highly in the olfactory bulb, choroid plexus and cerebellum. The distribution pattern agreed with the localization of PBR in the rodent brain. Infusion of kainic acid (KA: 1, 2.5 and 5 nmol) into the rat striatum resulted in neuroinflammation. In vitro and ex vivo ARG revealed that the radioactivity level of [ 11 C]AC-5216 was increased significantly in the KA-lesioned striatum compared to the non-lesioned striatum. Increasing the amount of KA infused into the striatum augmented radioactivity in the striatum as well as the cerebral cortex and hippocampus of the lesioned side. Treatment with a large amount of non-radioactive AC-5216 or PK11195 inhibited the binding of [ 11 C]AC-5216 and diminished the difference of radioactivity levels between the lesion and non-lesioned sides. These results demonstrated that [ 11 C]AC-5216 had high specific binding to PBR in the KA-lesioned rat brain. Thus, [ 11 C]AC-5216 is a promising PET ligand for imaging PBR in a brain with neuroinflammation.
- Published
- 2007
26. Expression of FasL and its interaction with Fas are mediated by c-Jun N-terminal kinase (JNK) pathway in 6-OHDA-induced rat model of Parkinson disease
- Author
-
Jing Pan, Zhiquan Wang, Sheng-Di Chen, Zhi Kun Sun, Yan xin Zhao, and Lei Jin
- Subjects
medicine.medical_specialty ,Fas Ligand Protein ,animal structures ,Dopamine ,Neurotoxins ,Apoptosis ,Substantia nigra ,Biology ,Fas ligand ,Rats, Sprague-Dawley ,Lesion ,Parkinsonian Disorders ,Internal medicine ,Neural Pathways ,medicine ,Animals ,fas Receptor ,Oxidopamine ,Pars compacta ,General Neuroscience ,c-jun ,JNK Mitogen-Activated Protein Kinases ,Brain ,Fas receptor ,Corpus Striatum ,Rats ,Up-Regulation ,Substantia Nigra ,Disease Models, Animal ,Endocrinology ,nervous system ,Sympatholytics ,Fas signaling pathway ,Cancer research ,Female ,Signal transduction ,medicine.symptom ,Signal Transduction - Abstract
Our previous studies and those of others have strongly suggested that c-Jun N-terminal kinase (JNK) signaling pathway plays a critical role in 6-hydroxydopamine (6-OHDA)-induced dopaminergic neuron injury in the substantia nigra. However, the downstream mechanism that accounts for the proapoptotic actions of JNK in 6-OHDA lesion remains to be investigated in detail. Fas, a member of the tumor necrosis factor receptor family with proapoptotic functions, was reported to be elevated within the striatum and substantia nigra pars compacta (SNc) of Parkinson's disease (PD) patients. In the present study, we examined the changes in the protein level of Fas ligand (FasL) and its interaction with Fas in a rat model of PD. We demonstrate that the expression of FasL and not Fas was increased after 6-OHDA lesion; additionally, the interaction of FasL and Fas was increased due to 6-OHDA lesion. This indicates that the 6-OHDA-induced activation of Fas signaling pathway is mediated by JNK and that FasL may be a promising target in the therapeutic approach for PD patients.
- Published
- 2007
27. Beta-amyloid toxicity and reversal in embryonic rat septal neurons
- Author
-
Karen M. Jarvis, Laura M. Mudd, Poincyane Assis-Nascimento, and Jeremy R. Montague
- Subjects
medicine.medical_specialty ,Programmed cell death ,Cell Survival ,Neurotoxins ,Basic fibroblast growth factor ,Apoptosis ,Biology ,Neuroprotection ,Drug Administration Schedule ,Article ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Insulin-Like Growth Factor II ,Internal medicine ,medicine ,Animals ,Insulin-Like Growth Factor I ,Cells, Cultured ,Neurons ,Amyloid beta-Peptides ,General Neuroscience ,Neurotoxicity ,Embryo, Mammalian ,medicine.disease ,Rats ,Chemically defined medium ,Neuroprotective Agents ,medicine.anatomical_structure ,Nerve growth factor ,Endocrinology ,Microscopy, Fluorescence ,chemistry ,Fibroblast Growth Factor 2 ,Septum Pellucidum ,Neuron - Abstract
Alzheimer's disease is characterized mainly by loss of neurons from the septal nucleus. In this study, neurons from the septal nucleus of the embryonic day 16 (E16) rat were grown in culture with a plane of astrocytes from the embryonic rat and in a defined medium in the absence of serum. Neurons were treated with beta-amyloid (Abeta: 0.1, 1 and 10 microM) on day in vitro (DIV) 1 and DIV 4 and fluorescent microscopy was used to measure survival and apoptosis following exposure of the treated cells on DIV 7. Reversal of neurotoxicity was studied using the potentially neuroprotective agents nerve growth factor (NGF, 100 ng/ml), basic fibroblast growth factor (bFGF, 5 ng/ml), insulin-like growth factors (IGF1 and IGF2, 10 ng/ml) and estrogen (10 nM), administered on DIV 4 and DIV 5, that is, subsequent to the Abeta (10 microM)-induced neurotoxicity. Abeta caused a significant decrease in survival at 10 microM, and a significant increase in apoptosis at 0.1 and 10 microM. IGF1, IGF2 and bFGF all caused a reversal of the Abeta-induced neurotoxic effect on survival while NGF and estrogen did not under these experimental conditions.
- Published
- 2007
28. Changes in pigment epithelium-derived factor expression following kainic acid induced cerebellar lesion in rat
- Author
-
Tomomi Sanagi, Haruki Yamada, and Takeshi Yabe
- Subjects
Male ,Calbindins ,medicine.medical_specialty ,Cerebellum ,Kainic acid ,Neurotoxins ,Purkinje cell ,Purkinje Cells ,chemistry.chemical_compound ,S100 Calcium Binding Protein G ,PEDF ,Cerebellar Diseases ,Internal medicine ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Gliosis ,Nerve Growth Factors ,RNA, Messenger ,Rats, Wistar ,Eye Proteins ,In Situ Hybridization, Fluorescence ,Serpins ,Kainic Acid ,Glial fibrillary acidic protein ,biology ,General Neuroscience ,Immunohistochemistry ,Rats ,Up-Regulation ,Endocrinology ,medicine.anatomical_structure ,nervous system ,Biochemistry ,chemistry ,Astrocytes ,Nerve Degeneration ,biology.protein ,Neuroglia ,Microglia ,Plant Lectins ,medicine.symptom ,Astrocyte - Abstract
Pigment epithelium-derived factor (PEDF) is a potent and broad-acting neurotrophic factor that protects various types of cultured neurons against glutamate excitotoxicity and induced apoptosis. The expression pattern and functions of PEDF in the central nervous system (CNS) remain largely undetermined. In this study, we analyzed the spatial and temporal expression of PEDF in normal and kainic acid (KA)-induced lesioned rat cerebellum using immunoblotting, immunohistochemistry and fluorescent in situ hybridization techniques. In normal rat cerebellum, PEDF protein and mRNA were mostly confined and co-localized with calbindin-positive cells in the Purkinje cell layer of the cerebellum, but not with glial fibrillary acidic protein (GFAP)-, 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNPase)-, and isolectin B4-positive cells. Injection of KA into the right cellebellum caused severe loss of calbindin-positive Purkinje neurons, and an increased number of GFAP-positive astrocytes and isolectin B4-positive microglia was observed on the ipsilateral side of the lesioned cerebellum. Although the PEDF level on the ipsilateral side of the cerebellum was dramatically decreased 2 days after KA treatment, significantly elevation of PEDF levels was observed at 7 days. In agreement with these results, PEDF protein and PEDF mRNA expression were co-localized with GFAP-positive reactive astrocytes in the ipsilateral side 7 days after KA treatment. Although the mechanism by which PEDF is induced in reactive astrocytes remains unclear, the increase in PEDF expression in injured brain may form part of a compensation mechanism against neuronal degeneration.
- Published
- 2007
29. Mutant SOD1G93A in bone marrow-derived cells exacerbates 3-nitropropionic acid induced striatal damage in mice
- Author
-
Liqun Yu, Qing-Yuan Huang, Robert J. Ferrante, and Jiang-Fan Chen
- Subjects
Male ,Pathology ,medicine.medical_specialty ,animal diseases ,medicine.medical_treatment ,Transgene ,Neurotoxins ,Intraperitoneal injection ,Central nervous system ,SOD1 ,Bone Marrow Cells ,Mice, Transgenic ,Biology ,Lesion ,Mice ,Superoxide Dismutase-1 ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Bone Marrow Transplantation ,Transplantation Chimera ,Superoxide Dismutase ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,nutritional and metabolic diseases ,Nitro Compounds ,Corpus Striatum ,nervous system diseases ,Mice, Inbred C57BL ,Oxidative Stress ,Huntington Disease ,medicine.anatomical_structure ,Mutation ,Nerve Degeneration ,Toxicity ,Systemic administration ,Bone marrow ,Propionates ,medicine.symptom ,Injections, Intraperitoneal - Abstract
3-Nitropropionic acid (3-NP), an irreversible inhibitor of succinate dehydrogenase, produces selective lesions in striatal neurons that resemble those observed in Huntington's disease neuropathology. In this study, we evaluated the role of peripheral bone marrow-derived cells (BMDCs) in the 3-NP-induced striatal damage by transplanting bone marrow cells with human SOD1 G93A mutation (mSOD1 G93A ) which induces amyotrophic lateral sclerosis through an unknown gain of toxicity and mitochondrial dysfunction. We assessed striatal damage after 3-NP treatment in the recipient C57BL/6 wild-type (WT) mice that received bone marrow cells from WT or mSOD1 G93A transgenic donor mice (WT → WT or mSOD G93A → WT). After intraperitoneal injection of 3-NP, six of the eight mSOD1 G93A → WT mice had bilateral striatal lesions while only one out of eight WT → WT mice had a striatal lesion. The lesion volume was significantly higher in the mSOD1 G93A → WT mice than in the WT → WT mice. However, following an intrastriatal injection of 3-NP, there was no significant difference in the lesion volumes between the WT → WT mice and mSOD1 G93A → WT mice. Thus, the exacerbation of 3-NP-induced striatal damage in mSOD G93A → WT mice was only seen after systemic administration of 3-NP, but not after intrastriatal injection. These results demonstrate that altered SOD1 activity (mSOD G93A ) in BMDCs affects striatal damage probably through a mechanism involving a systemic factor.
- Published
- 2007
30. Protective role of pentobarbital pretreatment for NMDA-R activated lipid peroxidation is derived from the synergistic effect on endogenous anti-oxidant in the hippocampus of rats
- Author
-
Akira Nakajima, Yuto Ueda, Keiko Nagatomo, and Taku Doi
- Subjects
Male ,Pentobarbital ,Free Radicals ,Neurotoxins ,Hippocampus ,Endogeny ,Pharmacology ,Hippocampal formation ,Receptors, N-Methyl-D-Aspartate ,Neuroprotection ,Antioxidants ,Lipid peroxidation ,chemistry.chemical_compound ,Excitatory Amino Acid Agonists ,medicine ,Animals ,Hypnotics and Sedatives ,Rats, Wistar ,General Neuroscience ,Rats ,Oxidative Stress ,Neuroprotective Agents ,chemistry ,Blood-Brain Barrier ,Hypoxia-Ischemia, Brain ,Anesthetic ,NMDA receptor ,Lipid Peroxidation ,Neuroscience ,medicine.drug - Abstract
We have attempted to explore the neuroprotective effectiveness of PBT by measuring anti-oxidant ability in the hippocampus of rats in a freely moving state. Anti-oxidant ability was examined utilizing the principle that blood–brain barrier-permeable nitroxide radicals (PCAM) injected i.p. lose their paramagnetism in an exponential decay correlated with anti-oxidant ability in the brain. The half-life of PCAM was used as the indicator of the hippocampal anti-oxidant ability. While the half-life was statistically prolonged when infused with 0.1 mM NMDA without PBT, the half-life was almost the same as in the control when infused with NMDA under anesthesia with PBT. In addition, the half-life under only PBT anesthesia was the shortest of all the groups. Our findings, therefore, suggested that PBT anesthesia not only suppresses NMDA-R activated free radical generation but also synergistically enhances the increased basal endogenous anti-oxidant ability in the hippocampus.
- Published
- 2007
31. Neuroprotective effect of carnosine on necrotic cell death in PC12 cells
- Author
-
Weiwei Hu, Haibin Dai, Yanying Fan, Zhong Chen, Yao Shen, and Qiuli Fu
- Subjects
Central Nervous System ,Programmed cell death ,N-Methylaspartate ,Neurotoxins ,Carnosine ,Histamine H1 receptor ,Pharmacology ,PC12 Cells ,Neuroprotection ,Necrosis ,chemistry.chemical_compound ,Excitatory Amino Acid Agonists ,medicine ,Animals ,Receptors, Histamine H1 ,Enzyme Inhibitors ,Neurons ,Pyrilamine ,Dose-Response Relationship, Drug ,Chemistry ,General Neuroscience ,Neurotoxicity ,Methylhistidines ,medicine.disease ,Histidine decarboxylase ,Mitochondria ,Rats ,Neuroprotective Agents ,Biochemistry ,Nerve Degeneration ,Histamine H1 Antagonists ,NMDA receptor ,Energy Metabolism ,Histamine - Abstract
The nervous tissue of many vertebrates, including humans, can synthesize beta-alanyl-L-histidine (carnosine). The biological functions of carnosine are still open to question, although several theories supported by strong experimental data have been proposed. The objective of this study was to examine the effects of carnosine on neurotoxicity in differentiated rat pheochromocytoma (PC12) cells. Neurotoxicity was induced by N-methyl-D-aspartate (NMDA), which caused time- and concentration-dependent cell death as measured by MTT and LDH assays. Pretreatment with carnosine significantly prevented the neurotoxicity in a concentration-dependent manner. The protective effect of carnosine was antagonized by the H1 receptor antagonist pyrilamine, but not by the H2 receptor antagonist cimetidine. In addition, alpha-fluoromethylhistidine, a histidine decarboxylase inhibitor, slightly reversed the protective action of carnosine. These results indicate that carnosine can effectively protect against NMDA-induced necrosis in PC12 cells, and its protection may in part be due to the activation of the postsynaptic histamine H1 receptor. The study suggests that carnosine may be an endogenous protective factor and calls for its further study as a new anti-excitotoxic agent.
- Published
- 2007
32. Spatiotemporal localization of injury potentials in DRG neurons during vincristine-induced axonal degeneration
- Author
-
Bruno Frazier, Surendra K. Ravula, Jonathan D. Glass, Min S. Wang, Maxine A. McClain, and Seneshaw Asress
- Subjects
Wallerian degeneration ,Neurotoxins ,Cell Culture Techniques ,Neural Conduction ,Degeneration (medical) ,Biology ,Article ,Membrane Potentials ,Rats, Sprague-Dawley ,Dorsal root ganglion ,Ganglia, Spinal ,medicine ,Animals ,Neurons, Afferent ,Axon ,Electrodes ,Cells, Cultured ,Dose-Response Relationship, Drug ,General Neuroscience ,Neurodegeneration ,Peripheral Nervous System Diseases ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Axons ,Rats ,Electrophysiology ,medicine.anatomical_structure ,nervous system ,Vincristine ,Soma ,Neuron ,Wallerian Degeneration ,Neuroscience ,Signal Transduction - Abstract
The distal to proximal degeneration of axons, or “dying back” is a common pattern of neuropathology in many diseases of the PNS and CNS. A long-standing debate has centered on whether this pattern of neurodegeneration is due to an insult to the cell body or to the axon itself, although it is likely that mechanisms are different for specific disease entities. We have addressed this question in a model system of vincristine-induced axonal degeneration. Here, we created a novel experimental apparatus combining a microfluidic divider with a multielectrode array substrate to allow for independent monitoring of injury-induced electrical activity from dorsal root ganglion (DRG) cell bodies and axons while isolating them into their own culture microenvironments. At specified doses, exposure of the cell body to vincristine caused neither morphological neurodegeneration nor persistent hyperexcitablility. In comparison, exposure of the distal axon to the same dose of vincristine first caused a decrease in the excitability of the axon and then axonal degeneration in a dying back pattern. Additionally, exposure of axons to vincristine caused an initial period of hyperexcitability in the cell bodies, suggesting that a signal is transmitted from the distal axon to the soma during the progression of vincristine-induced axonal degeneration. These data support the proposition that vincristine has a direct neurotoxic effect on the axon.
- Published
- 2007
33. Kainic acid induces early and transient autophagic stress in mouse hippocampus
- Author
-
Jianhua Zhang, Zuo-Lei Xie, Jun Lu, Yasuo Uchiyama, John J. Shacka, and Kevin A. Roth
- Subjects
Kainic acid ,Neurotoxins ,Excitotoxicity ,Mice, Transgenic ,Hippocampal formation ,Biology ,medicine.disease_cause ,Hippocampus ,Article ,Autophagy-Related Protein 5 ,Mice ,chemistry.chemical_compound ,Stress, Physiological ,Autophagy ,medicine ,Animals ,Ubiquitins ,PI3K/AKT/mTOR pathway ,Epilepsy ,Kainic Acid ,General Neuroscience ,Neurodegeneration ,medicine.disease ,Up-Regulation ,Cell biology ,Mice, Inbred C57BL ,Oncogene Protein v-akt ,chemistry ,Apoptosis ,Nerve Degeneration ,Neuron death ,Microtubule-Associated Proteins ,Neuroscience ,Biomarkers ,Molecular Chaperones - Abstract
Kainic acid (KA) treatment is a well-established model of hippocampal neuron death mediated in large part by KA receptor-induced excitotoxicity. KA-induced, delayed neuron death has been shown previously to follow the induction of seizures and exhibit characteristics of both apoptosis and necrosis. Growing evidence supports a role of autophagic stress-induced death of neurons in several in vitro and in vivo models of neuron death and neurodegeneration. However, whether autophagic stress also plays a role in KA-induced excitotoxicity has not been previously investigated. To examine whether KA alters the levels of proteins associated with or known to regulate the formation of autophagic vacuoles, we isolated hippocampal extracts from control mice and in mice following 2–16 h KA injection. KA induced a significant increase in the amount of LC3-II, a specific marker of autophagic vacuoles, at 4–6 h following KA, which indicates a transient induction of autophagic stress. Levels of autophagy-associated proteins including ATG5 (conjugated to ATG12), ATG6 and ATG7 did not change significantly after treatment with KA. However, ratios of phospho-mTOR/mTOR were elevated from 6 to 16 h, and ratios of phospho-Akt/Akt were elevated at 16 h following KA treatment, suggesting a potential negative feedback loop to inhibit further stimulation of autophagic stress. Together these data indicate the transient induction of autophagic stress by KA which may serve to regulate excitotoxic death in mouse hippocampus.
- Published
- 2007
34. Protection of vincristine-induced neuropathy by WldS expression and the independence of the activity of Nmnat1
- Author
-
Tadasuke Tsukiyama, Shigetsugu Hatakeyama, and Masashi Watanabe
- Subjects
Wallerian degeneration ,Neurite ,Blotting, Western ,Neurotoxins ,Retinoic acid ,Nerve Tissue Proteins ,Tretinoin ,Degeneration (medical) ,Biology ,Transfection ,E4B ,Cell Line ,Mice ,chemistry.chemical_compound ,491.4 ,NMNAT1 ,WldS ,Neurites ,medicine ,Animals ,Drug Interactions ,Nicotinamide-Nucleotide Adenylyltransferase ,Neurons ,General Neuroscience ,Cell Differentiation ,medicine.disease ,Fusion protein ,Cell biology ,chemistry ,Toxic injury ,Vincristine ,Nmnat1 ,Neuroscience - Abstract
The slow Wallerian degeneration protein (WldS), a fusion protein containing amino-terminal E4B and full-length nicotinamide mononucleotide adenylyltransferase 1 (Nmnat1), delays axon degeneration caused by physical damages, toxins and genetic mutations which result in patients being diagnosed with neurodegenerative diseases. It is still controversial whether the suppression of axonal degeneration by WldS is due to Nmnat1 or other portion. We generated WldS or Nmnat1-overexpressing Neuro2A cell lines, in which neuronal differentiation including neurite elongation can be induced by retinoic acid. The overexpression of WldS delayed the neurite degeneration by vincristine, whereas that of Nmnat1 did not delay it much. Taken together, Nmnat1 is considerably weaker than WldS for protection from toxic injury in vitro, suggesting that amino-terminal region of WldS is likely to be more significant for protection from axonal degeneration.
- Published
- 2007
35. Depletion of norepinephrine of the central nervous system Down-regulates the blood glucose level in d-glucose-fed and restraint stress models
- Author
-
Hong-Won Suh, Naveen Sharma, Soo-Hyun Park, Sung-Su Kim, and Jae-Ryeong Lee
- Subjects
0301 basic medicine ,Blood Glucose ,Central Nervous System ,Male ,Restraint, Physical ,medicine.medical_specialty ,Benzylamines ,medicine.medical_treatment ,Central nervous system ,Neurotoxins ,Down-Regulation ,DSP-4 ,Norepinephrine (medication) ,03 medical and health sciences ,chemistry.chemical_compound ,Norepinephrine ,0302 clinical medicine ,stomatognathic system ,Downregulation and upregulation ,D-Glucose ,Internal medicine ,medicine ,Neurotoxin ,Animals ,Injections, Spinal ,Injections, Intraventricular ,Catecholaminergic ,Mice, Inbred ICR ,business.industry ,General Neuroscience ,Insulin ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Glucose ,chemistry ,business ,Corticosterone ,030217 neurology & neurosurgery ,Stress, Psychological ,medicine.drug - Abstract
DSP-4[N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride] is a neurotoxin that depletes norepinephrine. The catecholaminergic system has been implicated in the regulation of blood glucose level. In the present study, the effect of DSP-4 administered intracerebroventricularly (i.c.v.) or intrathecally (i.t.) on blood glucose level was examined in d-glucose-fed and restraint stress mice models. Mice were pretreated once i.c.v. or i.t. with DSP-4 (10-40μg) for 3days, and d-glucose (2g/kg) was fed orally. Blood glucose level was measured 0 (prior to glucose feeding or restraint stress), 30, 60, and 120min after d-glucose feeding or restraint stress. The i.c.v. or i.t. pretreatment with DSP-4 attenuated blood glucose level in the d-glucose-fed model. Plasma corticosterone level was downregulated in the d-glucose-fed model, whereas plasma insulin level increased in the d-glucose-fed group. The i.c.v. or i.t. pretreatment with DSP-4 reversed the downregulation of plasma corticosterone induced by feeding d-glucose. In addition, the d-glucose-induced increase in plasma insulin was attenuated by the DSP-4 pretreatment. Furthermore, i.c.v. or i.t. pretreatment with DSP-4 reduced restraint stress-induced increases in blood glucose levels. Restraint stress increased plasma corticosterone and insulin levels. The i.c.v. pretreatment with DSP-4 attenuated restraint stress-induced plasma corticosterone and insulin levels. Our results suggest that depleting norepinephrine at the supraspinal and spinal levels appears to be responsible for downregulating blood glucose levels in both d-glucose-fed and restraint stress models.
- Published
- 2015
36. Modulation of BDNF and TrkB expression in rat hippocampus in response to acute neurotoxicity by diethyldithiocarbamate
- Author
-
Maria Rita Micheli, Moira Bazzucchi, Maria Assunta Laurenzi, R. Bova, and G. Grassi Zucconi
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,Neurotoxins ,Gene Expression ,Hippocampus ,Tropomyosin receptor kinase B ,Hippocampal formation ,Neurotrophic factors ,Internal medicine ,medicine ,Animals ,Receptor, trkB ,RNA, Messenger ,Rats, Wistar ,Brain-derived neurotrophic factor ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Chemistry ,Brain-Derived Neurotrophic Factor ,General Neuroscience ,Dentate gyrus ,Immunohistochemistry ,Rats ,Endocrinology ,Gene Expression Regulation ,nervous system ,Trk receptor ,biology.protein ,Ditiocarb ,Neurotrophin - Abstract
In this study, we examined the expression profile of brain-derived neurotrophic factor (BDNF) and its receptor TrkB in adult rat hippocampus following acute administration of diethyldithiocarbamate (DDTC), a neurotoxic compound which was previously shown to induce microglia activation and cell death. Semiquantitative RT-PCR analysis detected significant variations of BDNF mRNA levels in whole hippocampus homogenates, with a peak at 24 h after DDTC injection. Increased BDNF protein expression was demonstrated by immunohistochemistry in various hippocampal subfields. The most relevant increase was observed in the hilus of the dentate gyrus where BDNF levels at 120 h were found to be almost four times those of basal levels. Full-length TrkB (TrkB.FL) encoding mRNA was also shown to undergo an earlier increase in the hippocampus of DDTC-treated rats. TrkB immunostaining with an antibody binding both full-length and truncated (TrkB.T) isoforms was found to increase at 120 h in the hippocampal CA2 and CA3 regions. These results demonstrate that DDTC modulates the expression of BDNF and its receptor in the adult rat hippocampus and suggest a possible involvement of this neurotrophin in the protective response to DDTC-induced neuronal damage.
- Published
- 2006
37. Co-regulation of dopamine D1 receptor and uncoupling protein-2 expression in 3-nitropropionic acid-induced neurotoxicity: Neuroprotective role of l-carnitine
- Author
-
Antonino Amato, Nadia Salem, Ashraf Virmani, Zbigniew Binienda, Syed F. Ali, and Beata Przybyla
- Subjects
Male ,medicine.medical_specialty ,Neurotoxins ,Biology ,Ion Channels ,Body Temperature ,Mitochondrial Proteins ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Dopamine receptor D1 ,Dopamine ,Carnitine ,Internal medicine ,medicine ,Animals ,Drug Interactions ,Uncoupling Protein 2 ,RNA, Messenger ,Neurotransmitter ,Receptor ,Tyrosine hydroxylase ,Reverse Transcriptase Polymerase Chain Reaction ,Receptors, Dopamine D1 ,General Neuroscience ,Neurotoxicity ,Nitro Compounds ,medicine.disease ,Rats ,Neostriatum ,Disease Models, Animal ,Neuroprotective Agents ,Monoamine neurotransmitter ,Endocrinology ,Gene Expression Regulation ,chemistry ,Catecholamine ,Neurotoxicity Syndromes ,Propionates ,medicine.drug - Abstract
This study tested the hypothesis that the expression of uncoupling proteins (UCPs) and dopamine (DA) system genes is responsive to 3-nitropropionic acid (3-NPA) neurotoxic effects and to the neuroprotective effects of the mitochondrial enhancer, L-carnitine (LC), in the rat striatum. Inactivation of mitochondrial succinate dehydrogenase (SDH) by 3-NPA results in hypoxic brain damage. Hypoxic conditions induce uncoupling protein-2 (UCP-2). An increase in UCP-2 expression may lead to a decrease in production of reactive oxygen species (ROS) associated with energy depletion. However, this adaptive response can also lead to a reduction of ATP that may further contribute to energy deficit and mitochondrial dysfunction. Here, male adult Sprague-Dawley rats (n=5/group) were injected either with saline or 3-NPA at 30 mg/kg, s.c. alone or 30 min after pre-treatment with LC (100mg/kg, i.p.). Rectal temperature was monitored before treatment and 4h following 3-NPA administration. Animals were sacrificed 4h post-treatment. Total RNA was isolated from the striatum and transcripts of UCP-2, UCP-4 and UCP-5 genes, as well as genes related to dopamine metabolism, such as DA D(1) and D(2) receptors, tyrosine hydroxylase (TH), monoamine oxidase-B (MAO-B), and vesicular monoamine transporter-2 (VMAT-2), were measured using real-time reverse transcription polymerase chain reaction (RT-PCR). While core temperature decreased significantly in 3-NPA-treated rats, LC significantly inhibited the hypothermic effect of 3-NPA (p
- Published
- 2006
38. Ptychodiscus brevis toxin-induced depression of spinal reflexes involves 5-HT via 5-HT3 receptors modulated by NMDA receptor
- Author
-
Jitendra Singh, Rajesh Gupta, and Shripad B. Deshpande
- Subjects
Male ,Agonist ,Serotonin ,Spiperone ,medicine.medical_specialty ,Ketanserin ,medicine.drug_class ,Neurotoxins ,Biguanides ,Receptors, N-Methyl-D-Aspartate ,Internal medicine ,Reflex ,medicine ,Animals ,Dose-Response Relationship, Drug ,Chemistry ,General Neuroscience ,Oxocins ,Spinal cord ,Receptor antagonist ,Ondansetron ,Rats ,medicine.anatomical_structure ,Endocrinology ,Spinal Cord ,Depression, Chemical ,Dinoflagellida ,Dopamine Antagonists ,NMDA receptor ,Female ,Marine Toxins ,Serotonin Antagonists ,Receptors, Serotonin, 5-HT3 ,Spinal Nerve Roots ,medicine.drug ,Phenylbiguanide - Abstract
The involvement of 5-hydroxytryptaminergic (5-HT) system for the Ptychodiscus brevis toxin (PbTx)-induced depression of spinal reflexes was evaluated. The reflex potentials were recorded at ventral root by stimulating the corresponding dorsal root in neonatal rat spinal cord in vitro . Superfusion of PbTx (2.8–84 μM) depressed the monosynaptic (MSR) and polysynaptic (PSR) reflexes in a concentration-dependent manner. The depression of the reflexes was maximal with 84 μM of the toxin. Ondansetron (0.1 μM), a 5-HT 3 receptor antagonist, blocked the PbTx-induced depression of MSR and PSR. Spiperone (a 5-HT 2A antagonist) or ketanserin (5-HT 2A/2C antagonist and also at 5-HT 1B/1D ) failed to block the PbTx-induced depression of the reflexes. The 5-HT concentration of the cords was increased by four-fold after exposure to PbTx (28 μM) and the increase was not seen in the cords pretreated with dl -2 amino-5-phosphonovaleric acid (APV, a NMDA receptor antagonist). Superfusion of 5-HT or phenylbiguanide (PBG, a 5-HT 3 receptor agonist) also produced depression of the spinal reflexes in a concentration-dependent manner. The 5-HT-induced depression of reflexes was blocked by ondansetron but not by spiperone. The results demonstrate that the PbTx-induced depression of spinal reflexes involves 5-hydroxytryptamine via 5-HT 3 receptors modulated by NMDA receptor.
- Published
- 2006
39. Intragastric proteasome inhibition induces alpha-synuclein-immunopositive aggregations in neurons in the dorsal motor nucleus of the vagus in rats
- Author
-
Hideto Miwa, Ai Suzuki, Tomoyoshi Kondo, and Tomomi Kubo
- Subjects
Male ,Proteasome Endopeptidase Complex ,medicine.medical_specialty ,Retrograde Degeneration ,Neurotoxins ,Presynaptic Terminals ,Substantia nigra ,Biology ,Efferent Pathways ,Rats, Sprague-Dawley ,Parasympathetic nervous system ,Internal medicine ,medicine ,Animals ,Gliosis ,Enzyme Inhibitors ,Inclusion Bodies ,Medulla Oblongata ,General Neuroscience ,Stomach ,Parkinson Disease ,Vagus Nerve ,Environmental Exposure ,Environmental exposure ,Immunohistochemistry ,Rats ,Endocrinology ,medicine.anatomical_structure ,Dorsal motor nucleus ,nervous system ,alpha-Synuclein ,Proteasome inhibitor ,Lewy Bodies ,Microglia ,Neuron ,Neuron death ,Proteasome Inhibitors ,medicine.drug - Abstract
The neuropathological hallmark of idiopathic Parkinson's disease (PD) is dopaminergic neuron degeneration in the substantia nigra. However, it has been suggested that the neurodegenerative process initially may occur in the dorsal motor nucleus of the vagus (DMV). This implies that unidentified environmental toxins or neurotropic pathogens that is capable of passing the mucosal barrier of the gastrointestinal tract might affect the enteric nerve endings of the vagal neurons, possibly resulting in retrograde degeneration of the DMV. The present study aimed to evaluate the effects of proteasome inhibition of the intragastric nerve terminals of the DMV in rats. Following multiple injections of PSI, a selective proteasome inhibitor, or vehicle into the ventral wall of the stomach, the medulla oblongata was studied immunohistologically. In the DMV neurons of rats treated with PSI but not vehicle, alpha-synuclein-immunopositive intracytoplasmic inclusions and activated microglia were observed, predominantly in the left DMV. However, there was no significant loss of neurons. These results suggest that intragastric proteasome inhibition has a retrograde effect on DMV neurons but is insufficient to induce cell death, suggesting no causal linkage between inclusion body formation with proteasome inhibition and neuron death in the DMV. This might also implicate that Lewy body formation in the DMV in PD is possibly related to peroral invasion of environmental toxins that inhibit ubiquitin-proteasome system function.
- Published
- 2006
40. Activity of adenylyl cyclase and protein kinase A contributes to morphine-induced spinal apoptosis
- Author
-
Jeong-Ae Lim, Jianren Mao, Shuxing Wang, and Grewo Lim
- Subjects
Male ,MAPK/ERK pathway ,medicine.medical_specialty ,Programmed cell death ,MAP Kinase Signaling System ,Neurotoxins ,Apoptosis ,Biology ,Rats, Sprague-Dawley ,Adenylyl cyclase ,chemistry.chemical_compound ,Internal medicine ,Spinal Cord Dorsal Horn ,Cyclic AMP ,medicine ,Animals ,Drug Interactions ,Enzyme Inhibitors ,Protein kinase A ,Mitogen-Activated Protein Kinase 1 ,Neuronal Plasticity ,TUNEL assay ,Morphine ,Caspase 3 ,General Neuroscience ,Drug Tolerance ,Cyclic AMP-Dependent Protein Kinases ,Rats ,Enzyme Activation ,Posterior Horn Cells ,Endocrinology ,Nociception ,Terminal deoxynucleotidyl transferase ,chemistry ,Caspases ,Nerve Degeneration ,Adenylyl Cyclases - Abstract
Our previous study has shown that chronic morphine exposure induces neuronal apoptosis within the spinal cord dorsal horn; however, the mechanisms of morphine-induced apoptosis remain unclear. Here we examined whether adenylyl cyclase (AC) and protein kinase A (PKA) would play a role in this process. Intrathecal morphine regimen (10 μg, twice daily × 7 days) that resulted in antinociceptive tolerance induced spinal apoptosis as revealed by in situ terminal deoxynucleotidyl transferase (TdT)-UTP-biotin nick end labeling (TUNEL). The TUNEL-positive cells were detected primarily in the superficial laminae of the spinal cord dorsal horn, which was associated with an increase in the expression of activated caspase-3 and mitogen-activated protein kinase (MAPK) within the same spinal region. Co-administration of morphine with a broad AC inhibitor (ddA), a PKA inhibitor (H89), or a MAPK inhibitor (PD98059) substantially reduced the number of TUNEL-positive cells, as compared with the morphine alone group. The results indicate that the spinal AC and PKA pathway through intracellular MAPK may be contributory to the cellular mechanisms of morphine-induced apoptosis.
- Published
- 2005
41. Amburoside A, a glucoside from Amburana cearensis, protects mesencephalic cells against 6-hydroxydopamine-induced neurotoxicity
- Author
-
H.V. Nobre Júnior, Geanne M. A. Cunha, Manoel Odorico de Moraes, Cláudia Pessoa, R.A. Oliveira, Edilberto R. Silveira, K. M. Canuto, G.S.B. Viana, and Luzia Kalyne Almeida Moreira Leal
- Subjects
Thiobarbituric acid ,Neurotoxins ,In Vitro Techniques ,Pharmacology ,Antioxidants ,Nitric oxide ,chemistry.chemical_compound ,Glucosides ,Mesencephalon ,Pregnancy ,TBARS ,Animals ,Drug Interactions ,MTT assay ,Viability assay ,Rats, Wistar ,Nitrite ,Oxidopamine ,Neurons ,Hydroxydopamine ,biology ,Plant Extracts ,Chemistry ,General Neuroscience ,Fabaceae ,biology.organism_classification ,Rats ,Neuroprotective Agents ,Biochemistry ,Amburana cearensis ,Plant Bark ,Sympatholytics ,Female - Abstract
This study evaluates the potential neuroprotective properties of amburoside A, a glucoside isolated from Amburana cearensis, on rat mesencephalic cell cultures exposure to the neurotoxin 6-hydroxydopamine (6-OHDA). The parameters determined were cell viability by the 3[4,5-dimethylthiazole-2-il]-2,5-diphenyltetrazolium bromide (MTT) method, nitric oxide (NO) and free radical formation by the measurement of nitrite concentration and thiobarbituric acid reacting substance (TBARS) formation as an indication of cellular lipid peroxidation. The results showed that AMB was less effective as a curative agent in the MTT assay, since its addition after 6-OHDA did not reverse the neurotoxin's effect, except at the highest concentration (AMB, 100 microg/ml). Similarly, the higher nitrite levels observed after exposure of the cells to 6-OHDA were only partially reversed by AMB, at this highest concentration. However, when AMB (0.5, 1, 10 and 100 microg/ml) was added before the toxin, it appeared to protect neuronal cells against 6-OHDA toxicity in a concentration-dependent manner, as shown by MTT assay. AMB also prevented free radical formation indicated by the increased nitrite concentration induced by 6-OHDA. Cells exposed to 6-OHDA showed a 3.4 times increase in TBARS concentration as compared to controls, and this effect was inhibited from 24% up to 64% by AMB (0.1-100 microg/ml), indicative of a neuroprotective effect. In conclusion, we show that AMB, acting as an antioxidant compound, presents a significant neuroprotective effect, suggesting that this compound could provide benefits as a therapeutic agent in neurodegenerative disease such as Parkinson's.
- Published
- 2005
42. Neuronal markers expression of NGF-primed bone marrow cells (BMCs) transplanted in the brain of 6-hydroxydopamine and ibotenic acid lesioned littermate mice
- Author
-
Luigi Aloe and Viviana Triaca
- Subjects
Male ,medicine.medical_specialty ,Tyrosine 3-Monooxygenase ,Cell Survival ,Blotting, Western ,Neurotoxins ,Bone Marrow Cells ,Tropomyosin receptor kinase A ,Biology ,Mice ,Pregnancy ,Internal medicine ,Nerve Growth Factor ,medicine ,Animals ,Drug Interactions ,Receptor, trkA ,Oxidopamine ,Ibotenic Acid ,Bone Marrow Transplantation ,Hydroxydopamine ,Dose-Response Relationship, Drug ,Tyrosine hydroxylase ,General Neuroscience ,Immunohistochemistry ,Choline acetyltransferase ,Transplantation ,Nerve growth factor ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,Bromodeoxyuridine ,Gene Expression Regulation ,nervous system ,Brain Injuries ,Cholinergic ,Female ,Bone marrow - Abstract
In the present study, we aim to show that non-adherent bone marrow cells (BMCs) express TrkA, the nerve growth factor (NGF) receptor, and that addition of NGF promotes the survival and neuronal commitment of BMC transplanted into the experimentally injured brain of littermates mice. Immunohistochemical analysis revealed that transplanted BMCs express tyrosine hydroxylase (TH) in proximity of the damaged dopaminergic tissues and choline acetyltransferase (ChAT) in the lesioned cholinergic regions. These results suggest that NGF supports the survival and differentiation of uncommitted BMCs and concurs with other local environmental signals to promote the expression of neuronal markers in these cells. The possible functional significance of these observations will be discussed.
- Published
- 2005
43. Kanamycin ototoxicity in glutamate transporter knockout mice
- Author
-
Jun Hyodo, Kiyofumi Gyo, Yoshitaka Shimizu, Nobuhiro Hakuba, and Masafumi Taniguchi
- Subjects
medicine.medical_specialty ,Synaptic cleft ,Amino Acid Transport System X-AG ,Hearing Loss, Sensorineural ,Neurotoxins ,Glutamic Acid ,Biology ,Synaptic Transmission ,Mice ,Ototoxicity ,Kanamycin ,Internal medicine ,Evoked Potentials, Auditory, Brain Stem ,otorhinolaryngologic diseases ,medicine ,Animals ,Cochlea ,Antibacterial agent ,Mice, Knockout ,Hair Cells, Auditory, Inner ,General Neuroscience ,Glutamate receptor ,Auditory Threshold ,medicine.disease ,medicine.anatomical_structure ,Endocrinology ,Nerve Degeneration ,Knockout mouse ,sense organs ,Hair cell ,Neuroscience ,medicine.drug - Abstract
Glutamate-aspartate transporter (GLAST), a powerful glutamate uptake system, removes released glutamate from the synaptic cleft and facilitates the re-use of glutamate as a neurotransmitter recycling system. Aminoglycoside-induced hearing loss is mediated via a glutamate excitotoxic process. We investigated the effect of aminoglycoside ototoxicity in GLAST knockout mice using the recorded auditory brainstem response (ABR) and number of hair cells in the cochlea. Kanamycin (100 mg/mL) was injected directly into the posterior semicircular canal of mice. Before the kanamycin treatment, there was no difference in the ABR threshold average between the wild-type and knockout mice. Kanamycin injection aggravated the ABR threshold in the GLAST knockout mice compared with the wild-type mice, and the IHC degeneration was more severe in the GLAST knockout mice. These findings suggest that GLAST plays an important role in preventing the degeneration of inner hair cells in aminoglycoside ototoxicity.
- Published
- 2005
44. Spinal neurons involved in the generation of at-level pain following spinal injury in the rat
- Author
-
Patrick W. Mantyh, Charles J. Vierck, Douglas A. Lappi, Chen Guang Yu, and Robert P. Yezierski
- Subjects
Saporin ,Neurotoxins ,Population ,Excitotoxicity ,Pain ,Substance P ,medicine.disease_cause ,chemistry.chemical_compound ,Animals ,Medicine ,Quisqualic acid ,education ,N-Glycosyl Hydrolases ,Spinal cord injury ,Spinal Cord Injuries ,Plant Proteins ,Skin ,education.field_of_study ,Behavior, Animal ,biology ,business.industry ,Immunotoxins ,General Neuroscience ,Receptors, Neurokinin-1 ,Spinal cord ,medicine.disease ,Grooming ,Immunohistochemistry ,Saporins ,Rats ,Posterior Horn Cells ,Disease Models, Animal ,medicine.anatomical_structure ,Spinal Cord ,chemistry ,Anesthesia ,Ribosome Inactivating Proteins, Type 1 ,Self Mutilation ,biology.protein ,Neuron ,business - Abstract
Using a conjugate of substance P and the ribosome-inactivating protein saporin, neurons expressing the neurokinin-1 receptor in lamina I of the spinal cord were targeted to determine their role in the expression of a spontaneous pain behavior following intraspinal injections of quisqualic acid in the rat. Treatment was carried out at the time of injury in order to prevent the onset of the behavior, and following onset in order to evaluate the potential clinical utility of this intervention. Treatment at the time of injury resulted in significant decreases in onset-time and severity of pain behavior, while treatment at the time of onset led to a significant reduction of the spontaneous self-directed behavior. The results suggest that the substrate for at-level pain following spinal cord injury includes a population of spinal neurons expressing the neurokinin-1 receptor in the superficial laminae of the spinal cord.
- Published
- 2004
45. Protective effects of S-nitrosoglutathione against neurotoxicity of 3-nitropropionic acid in rat
- Author
-
Ya Ting Yang, Ding I. Yang, and Tzyh Chwen Ju
- Subjects
Antioxidant ,Cell Survival ,medicine.medical_treatment ,Neurotoxins ,Tetrazolium Salts ,Pharmacology ,Mitochondrion ,Biology ,medicine.disease_cause ,Neuroprotection ,Nitric oxide ,S-Nitrosoglutathione ,chemistry.chemical_compound ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Drug Interactions ,Cells, Cultured ,Cerebral Cortex ,Neurons ,Analysis of Variance ,Cell Death ,Dose-Response Relationship, Drug ,L-Lactate Dehydrogenase ,General Neuroscience ,Neurotoxicity ,Glutathione ,Embryo, Mammalian ,Nitro Compounds ,medicine.disease ,Immunohistochemistry ,Rats ,Thiazoles ,Neuroprotective Agents ,chemistry ,Biochemistry ,Propionates ,Microtubule-Associated Proteins ,Oxidative stress - Abstract
Mitochondrial dysfunction and oxidative stress are often linked to various neurodegenerative disorders including ischemic stroke and Huntington's disease (HD). S-Nitrosoglutathione (GSNO) is an endogenous nitric oxide carrier recently identified as a potent antioxidant capable of neutralizing oxidative stress. In the present study, we explore the neuroprotective effects of GSNO against metabolic insults induced by 3-nitropropionic acid (3-NP), a mitochondrial complex II inhibitor commonly used as a pharmacological model for HD, in primary culture of fetal rat cortical and striatal neurons. Application of GSNO (1-5 microM) substantially reduced neuronal loss caused by 3-NP (1-5 mM) exposure based on MTT reduction, lactate dehydrogenase (LDH) release, and Hoechst staining assays. The protective effect of GSNO appeared to be more potent than N-acetyl-l-cysteine (NAC), a glutathione precursor, at the same concentrations. These results suggest that manipulation of GSNO metabolism may exert protective effects against mitochondrial dysfunction often observed in neurodegenerative disorders.
- Published
- 2004
46. Synergism between tumor necrosis factor-α and H2O2 enhances cell damage in rat PC12 cells
- Author
-
Saleh Abu-Raya, Esther Shohami, and Victoria Trembovler
- Subjects
Programmed cell death ,medicine.medical_treatment ,Neurotoxins ,Biology ,Pharmacology ,medicine.disease_cause ,PC12 Cells ,Antioxidants ,Dinoprostone ,medicine ,Animals ,Dronabinol ,Cell damage ,chemistry.chemical_classification ,Reactive oxygen species ,Cell Death ,L-Lactate Dehydrogenase ,Tumor Necrosis Factor-alpha ,General Neuroscience ,Drug Synergism ,Hydrogen Peroxide ,medicine.disease ,Rats ,Oxidative Stress ,Cytokine ,chemistry ,Immunology ,Toxicity ,Tumor necrosis factor alpha ,Reactive Oxygen Species ,Oxidative stress ,Prostaglandin E - Abstract
Tumor necrosis factor-alpha (TNFalpha) is harmful in the early phase and beneficial in the long-term phase after brain injury. Reactive oxygen species (ROS) are among the most toxic mediators activated by injury. We speculate that part of the TNFalpha toxicity is mediated by its synergism with ROS. Thus, toxicity of TNFalpha and ROS, alone or together, were studied in PC12 cells. PC12 cells were exposed for 18 h to TNFalpha (0-100 ng/ml), to H2O2 (1-300 microM) or to both, each at sub-toxic concentrations. Lactic dehydrogenase release, prostaglandin E2 accumulation and morphology indicated cell death and stress response. TNFalpha toxicity was seen at50 ng/ml, and that of H2O2 at150 microM, however, when together, sub-lethal levels (25 ng/ml TNFalpha and 30 microM H2O2) induced toxicity. Dexanabinol, an N-methyl-D-aspartate antagonist with antioxidant and anti-TNFalpha properties, completely rescued the cells. These findings corroborate our hypothesis on the cooperative toxicity exerted by TNFalpha and ROS after brain injury.
- Published
- 2003
47. N-Methyl-d-aspartate receptor subunit expression and phosphorylation following excitotoxic spinal cord injury in rats
- Author
-
Federico M Perez, Christopher D. King, Robert M. Caudle, Robert P. Yezierski, and Chen Guang Yu
- Subjects
medicine.medical_specialty ,Protein subunit ,Neurotoxins ,Central nervous system ,Synaptic Membranes ,Excitotoxicity ,Biology ,medicine.disease_cause ,Receptors, N-Methyl-D-Aspartate ,Synaptic Transmission ,Internal medicine ,Serine ,medicine ,Animals ,Phosphorylation ,Spinal cord injury ,Spinal Cord Injuries ,Neurons ,musculoskeletal, neural, and ocular physiology ,General Neuroscience ,Glutamate receptor ,Quisqualic Acid ,Spinal cord ,medicine.disease ,Grooming ,Endocytosis ,Rats ,Disease Models, Animal ,Protein Transport ,medicine.anatomical_structure ,Endocrinology ,Spinal Cord ,nervous system ,Immunology ,NMDA receptor ,sense organs - Abstract
The role of NMDA receptor expression and post-translational modification in the pathological and behavioral consequences of injury were examined in rats receiving spinal injections of quisqualate. Spinal cords were removed 3 days following the development of excessive grooming behavior or, if the spontaneous pain-like behavior was not observed, 13 days following injections. Western blots from the spinal tissue demonstrated that non-grooming animals had elevated protein levels of the NR1 subunit of the NMDA receptor. These subunits did not demonstrate an enhanced level of phosphorylation. NR1 protein in grooming rats was not elevated, but there was a significant increase in NR1 serine phosphorylation. These findings suggest that excitotoxic lesions of the spinal cord induce both NR1 expression and NR1 serine phosphorylation. However, the injury-induced excessive grooming behavior is only associated with phosphorylation of the NR1 subunit.
- Published
- 2003
48. Role of glutamate receptors and an on-going protein synthesis in the regulation of phosphorylation of Ca2+/calmodulin-dependent protein kinase II in the CA3 hippocampal region in mice administered with kainic acid intracerebroventricularly
- Author
-
Hong-Won Suh, Han-Kyu Lee, Ki-Jung Han, Eun-Jung Han, and Seong-Soo Choi
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Kainic acid ,Neurotoxins ,Cycloheximide ,Hippocampus ,Mice ,chemistry.chemical_compound ,Internal medicine ,medicine ,Protein biosynthesis ,Animals ,Phosphorylation ,Receptor ,Injections, Intraventricular ,Neurons ,Protein Synthesis Inhibitors ,Mice, Inbred ICR ,Kainic Acid ,General Neuroscience ,Glutamate receptor ,Up-Regulation ,Endocrinology ,Receptors, Glutamate ,chemistry ,Calcium-Calmodulin-Dependent Protein Kinases ,CNQX ,NMDA receptor ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Excitatory Amino Acid Antagonists - Abstract
In an immunohistochemical study, kainic acid (KA, 0.1 microg) administered intracerebroventricularly (i.c.v.) dramatically increased the expression of Ca2+/calmodulin-dependent protein kinase II (CaMK II) and the phosphorylation of CaMK II (p-CaMK II) in the CA3 hippocampal region of mice. Pre-treatment with cycloheximide (a protein synthesis inhibitor; 200 mg/kg) intraperitoneally prevented the expression of CaMK II and phosphorylation of CaMK II induced by KA. In addition, pre-treatment with MK-801 ((5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine; an NMDA receptor blocker, 1 microg, i.c.v.) or CNQX (6-cyano-7-nitroquinoxaline-2,3-dione; a non-NMDA receptor blocker, 0.5 microg, i.c.v.) attenuated the p-CaMK II, but not CaMK II, expression induced by KA. Our results suggest that KA administered supraspinally induces CaMK II and the phosphorylation of CaMK II expression in the CA3 hippocampal region, for which an on-going protein synthesis is needed. Furthermore, both NMDA and non-NMDA receptors appear to be involved in supraspinally administered KA-induced phosphorylation of CaMK II.
- Published
- 2003
49. Denervation of the locus coeruleus projections by treatment with the selective neurotoxin DSP-4 [N (2-chloroethyl)-N-ethyl-2-bromobenzylamine] reduces dopamine release potential in the nucleus accumbens shell in conscious rats
- Author
-
Marika Eller, Ivo Kolts, Riina Häidkind, Lars Oreland, Jaanus Harro, and Toomas Kivastik
- Subjects
Male ,Benzylamines ,Microdialysis ,medicine.medical_specialty ,Dopamine ,Neurotoxins ,Prefrontal Cortex ,DSP-4 ,Nucleus accumbens ,Biology ,Nucleus Accumbens ,chemistry.chemical_compound ,Internal medicine ,Neural Pathways ,Electrochemistry ,medicine ,Animals ,Neurotoxin ,Rats, Wistar ,Neurotransmitter ,Chromatography, High Pressure Liquid ,General Neuroscience ,Denervation ,Rats ,Endocrinology ,chemistry ,Catecholamine ,Locus coeruleus ,Locus Coeruleus ,Neuroscience ,medicine.drug - Abstract
Pretreatment with DSP-4, a neurotoxin highly selective for the locus coeruleus (LC) noradrenergic projections, 2 weeks before in vivo microdialysis in conscious rats had no effect on baseline extracellular dopamine (DA) levels in the nucleus accumbens shell, but reduced dose-dependently the dopamine response to depolarisation induced by 50 mM KCl. DA metabolism in the frontal cortex, as measured ex vivo, was increased in animals treated with a low (10 mg/kg) but not with a high dose (50 mg/kg) of DSP-4, possibly indicating an increased sensitivity to stress in these animals and thus suggesting differential regulation of DA in the forebrain by the LC lesions. The reduced DA release potential in the nucleus accumbens after DSP-4 treatment suggests that weakening of the LC input to DA nerve cells contributes to motivational deficits.
- Published
- 2002
50. Acetyl-l-carnitine shows neuroprotective and neurotrophic activity in primary culture of rat embryo motoneurons
- Author
-
S Larini, V Muzio, Paolo Bigini, Tiziana Mennini, and Claudio Pasquali
- Subjects
medicine.medical_specialty ,Neurotoxins ,Kainate receptor ,Biology ,Neuroprotection ,Choline O-Acetyltransferase ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Fetus ,Neurotrophic factors ,Internal medicine ,medicine ,Animals ,Choline ,Receptor ,Cells, Cultured ,Nootropic Agents ,Motor Neurons ,General Neuroscience ,Amyotrophic Lateral Sclerosis ,Receptor Protein-Tyrosine Kinases ,Choline acetyltransferase ,Rats ,Neuroprotective Agents ,Endocrinology ,nervous system ,Biochemistry ,chemistry ,biology.protein ,NMDA receptor ,Acetylcarnitine ,Neurotrophin - Abstract
We evaluated the role of acetyl-L-carnitine (ALCAR) in protecting primary motoneuron cultures exposed to excitotoxic agents or serum-brain derived neurotrophic factor (BDNF) deprived. To exclude that ALCAR works as a metabolic source, we compared its effects with those of L-carnitine (L-CAR), that seems to have no neurotrophic effect. A concentration of 10 mM ALCAR, but not L-CAR, significantly reduced the toxic effect of 50 microM N-methyl-D-aspartate (NMDA, % viability: NMDA 45.4+/-2.80, NMDA+ALCAR 90.8+/-11.8; P
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.