Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Exp Brain Res ; 240(11): 2981-2988, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36181544

ABSTRACT

The mother-child relationship of newborns plays an essential role in the development of the central nervous system, and an inadequate relationship, such as mother-child separation, can cause deficits of mental function in adulthood. However, insufficient research has examined the effects of foster mothers. We assigned some neonatal rats to one of two foster mothers: one that was lactating and feeding her first litter (FL group) and one that had one previous experience of childbirth and feeding but no current litter (FE group). Other pups were raised by their own mother (OM group) or subjected to maternal separation (MS group). Pups were placed with the foster mother (FL and FE groups) or separated from their mother (MS group) for 3 h/day on postnatal days 1-20. At age 6 weeks, each group was divided into two subgroups, one with 30 min of acute restraint stress loading (FL-R, FE-R, OM-R, and MS-R) and one without it (FL, FE, OM, and MS). Then, we compared the density of corticotropin-releasing factor-immunoreactive (CRF-ir) neurons in the central amygdaloid nucleus (CeA). The density of CRF-ir neurons in the CeA was significantly lower in the FL-R and MS-R subgroups than in the FL and MS subgroups, respectively. The results suggest that differences in care received during the neonatal period affect maturation of CRF neurons in the CeA and may have negative effects on the synthesis and release of CRF.


Subject(s)
Central Amygdaloid Nucleus , Corticotropin-Releasing Hormone , Female , Rats , Animals , Humans , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Maternal Deprivation , Mothers , Lactation , Neurons/metabolism
2.
Protein Pept Lett ; 27(5): 432-446, 2020.
Article in English | MEDLINE | ID: mdl-31652112

ABSTRACT

BACKGROUND: Intermediate Filaments (IFs) are major constituents of the cytoskeletal systems in animal cells. OBJECTIVE: To gain insights into the structure-function relationship of invertebrate cytoplasmic IF proteins, we characterized an IF protein from the platyhelminth, Dugesia japonica, termed Dif-1. METHODS: cDNA cloning, in situ hybridization, immunohistochemical analysis, and IF assembly experiments in vitro using recombinant Dif-1, were performed for protein characterization. RESULTS: The structure deduced from the cDNA sequence showed that Djf-1 comprises 568 amino acids and has a tripartite domain structure (N-terminal head, central rod, and C-terminal tail) that is characteristic of IF proteins. Similar to nuclear IF lamins, Djf-1 contains an extra 42 residues in the coil 1b subdomain of the rod domain that is absent from vertebrate cytoplasmic IF proteins and a nuclear lamin-homology segment of approximately 105 residues in the tail domain; however, it contains no nuclear localization signal. In situ hybridization analysis showed that Djf-1 mRNA is specifically expressed in cells located within the marginal region encircling the worm body. Immunohistochemical analysis showed that Djf-1 protein forms cytoplasmic IFs located close to the microvilli of the cells. In vitro IF assembly experiments using recombinant proteins showed that Djf-1 alone polymerizes into IFs. Deletion of the extra 42 residues in the coil 1b subdomain resulted in the failure of IF formation. CONCLUSION: Together with data from other histological studies, our results suggest that Djf- 1 is expressed specifically in anchor cells within the glandular adhesive organs of the worm and that Djf-1 IFs may play a role in protecting the cells from mechanical stress.


Subject(s)
Intermediate Filament Proteins/genetics , Planarians/chemistry , Planarians/genetics , Recombinant Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Evolution, Molecular , Gene Expression Regulation , Humans , Intermediate Filaments/metabolism , Lamins/genetics , Mutation , Protein Conformation , RNA, Messenger/metabolism , Structure-Activity Relationship
4.
Mol Pain ; 14: 1744806918796057, 2018.
Article in English | MEDLINE | ID: mdl-30152258

ABSTRACT

Extracellular regulated protein kinase (ERK) pathway activation in astrocytes and neurons has been reported to be critical for neuropathic pain development after chronic constriction injury. TGN-020 was found to be the most potent aquaporin 4 inhibitor among the agents studied. The present study aimed to assess whether the inhibition of aquaporin 4 had an analgesic effect on neuropathic pain and whether the inhibition of astrocytic activation and ERK pathway was involved in the analgesic effect of TGN-020. We thus found that TGN-020 upregulated the threshold of thermal and mechanical allodynia, downregulated the expression of interleukin-1ß, interleukin-6, and tumor necrosis factor-α, attenuated the astrocytic activation and suppressed the activation of mitogen-activated protein kinase pathways in the spinal dorsal horn and dorsal root ganglion. Additionally, TGN-020 suppressed ERK phosphorylation in astrocytes and neurons after injury. The findings suggested that the analgesic effects of TGN-020 in neuropathic pain were mediated mainly by the downregulation of chronic constriction injury-induced astrocytic activation and inflammation, which is via the inhibition of ERK pathway in the spinal dorsal horn and dorsal root ganglion.


Subject(s)
Analgesics/therapeutic use , MAP Kinase Signaling System/drug effects , Neuralgia/drug therapy , Niacinamide/analogs & derivatives , Thiadiazoles/therapeutic use , Animals , Aquaporin 4/antagonists & inhibitors , Aquaporin 4/metabolism , Disease Models, Animal , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Niacinamide/therapeutic use , Pain Threshold/drug effects , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
5.
J Neuroinflammation ; 14(1): 67, 2017 03 29.
Article in English | MEDLINE | ID: mdl-28356158

ABSTRACT

BACKGROUND: Astrocyte activation is a hallmark of traumatic brain injury resulting in neurological dysfunction or death for an overproduction of inflammatory cytokines and glial scar formation. Both the silent mating type information (Sirt1) expression and mitogen-activated protein kinase (MAPK) signal pathway activation represent a promising therapeutic target for several models of neurodegenerative diseases. We investigated the potential effects of Sirt1 upregulation and MAPK pathway pharmacological inhibition on astrocyte activation in vitro and in vivo. Moreover, we attempted to confirm the underlying interactions between Sirt1 and MAPK pathways in astrocyte activation after brain injury. METHODS: The present study employs an interleukin-1ß (IL-1ß) stimulated primary cortical astrocyte model in vitro and a nigrostriatal pathway injury model in vivo to mimic the astrocyte activation induced by traumatic brain injury. The activation of GFAP, Sirt1, and MAPK pathways were detected by Western blot; astrocyte morphological hypertrophy was assessed using immunofluorescence staining; in order to explore the neuroprotective effect of regulation Sirt1 expression and MAPK pathway activation, the motor and neurological function tests were assessed after injury. RESULTS: GFAP level and morphological hypertrophy of astrocytes are elevated after injury in vitro or in vivo. Furthermore, the expressions of phosphorylated extracellular regulated protein kinases (p-ERK), phosphorylated c-Jun N-terminal kinase (p-JNK), and phosphorylated p38 activation (p-p38) are upregulated, but the Sirt1 expression is downregulated. Overexpression of Sirt1 significantly increases the p-ERK expression and reduces the p-JNK and p-p38 expressions. Inhibition of ERK, JNK, or p38 activation respectively with their inhibitors significantly elevated the Sirt1 expression and attenuated the astrocyte activation. Both the overproduction of Sirt1 and inhibition of ERK, JNK, or p38 activation can alleviate the astrocyte activation, thereby improving the neurobehavioral function according to the modified neurological severity scores (mNSS) and balance latency test. CONCLUSIONS: Thus, Sirt1 plays a protective role against astrocyte activation, which may be associated with the regulation of the MAPK pathway activation induced by brain injury in vitro and in vivo.


Subject(s)
Astrocytes/metabolism , Brain Injuries/metabolism , Mitogen-Activated Protein Kinases/metabolism , Sirtuin 1/metabolism , Animals , Animals, Newborn , Astrocytes/pathology , Brain Injuries/pathology , Cells, Cultured , Humans , Male , Mice , Protein Binding/physiology , Sirtuin 1/genetics
6.
Exp Brain Res ; 235(4): 1053-1062, 2017 04.
Article in English | MEDLINE | ID: mdl-28070623

ABSTRACT

It is known that the median preoptic nucleus (POMe) sends dense projections to the subfornical organ (SFO). However, the functional significance of these projections have not been well discussed. In this electron microscopic study, we investigated the presence of synapses between POMe-derived axon terminals and SFO neurons that project to the paraventricular hypothalamic nucleus (PVN). After injection of a retrograde tracer, wheat germ agglutinin-conjugated horseradish peroxidase-colloidal gold complex, into the PVN, many labeled neurons were found in the SFO. In contrast, after injection of an anterograde tracer, biotinylated dextran amine, in the POMe, abundant labeled axon varicosities were observed in the SFO. Using electron microscopy, synapses were identified between retrogradely labeled dendrites and cell bodies, and anterogradely labeled axon terminals, indicating that POMe neurons innervate SFO neurons projecting to the PVN. The possibility that POMe neurons play multiple roles in the neuronal circuit responsible for vasopressin release and/or cardiovascular regulation is also discussed.


Subject(s)
Neurons/physiology , Paraventricular Hypothalamic Nucleus/cytology , Preoptic Area/cytology , Subfornical Organ/cytology , Synapses/physiology , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Dextrans/metabolism , Gold Colloid/metabolism , Male , Microinjections , Microscopy, Immunoelectron , Neural Pathways/physiology , Neurons/metabolism , Neurons/ultrastructure , Rats , Rats, Sprague-Dawley , Synapses/ultrastructure , Wheat Germ Agglutinins/metabolism
7.
Brain Res ; 1654(Pt A): 1-8, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27769789

ABSTRACT

Nigrostriatal pathway injury is one of the traumatic brain injury models that usually lead to neurological dysfunction or neuron necrosis. Resveratrol-induced benefits have recently been demonstrated in several models of neuronal degeneration diseases. However, the protective properties of resveratrol against neurodegeneration have not been explored definitely. Thus, we employ the nigrostriatal pathway injury model to mimic the insults on the brain. Resveratrol decreased the p-ERK expression and increased the p-JNK expression compared to the DMSO group, but not alter the p38 MAPK proteins around the lesion site by Western blot. Prior to the injury, mice were infused with resveratrol intracerebroventricularly with or without JNK-IN-8, a specific c-JNK pathway inhibitor for JNK1, JNK2 and JNK4. The study assessed modified improved neurological function score (mNSS) and beam/walking test, the level of inflammatory cytokines IL-1ß, IL-6 and TNF-α, and striatal expression of Bax and Bcl-2 proteins associated with neuronal apoptosis. The results revealed that resveratrol exerted a neuroprotective effect as shown by the improved mNSS and beam latency, anti-inflammatory effects as indicated by the decreased level of IL-1ß, TNF-α and IL-6. Furthermore, resveratrol up-regulated the protein expression of p-JNK and Bcl-2, down-regulated the expression of Bax and the number of Fluoro-Jade C (FJC) positive neurons. However, these advantages of resveratrol were abolished by JNK-IN-8 treatment. Overall, we demonstrated that resveratrol treatment attenuates the nigrostriatal pathway injury-induced neuronal apoptosis and inflammation via activation of c-JNK signaling.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Corpus Striatum/drug effects , MAP Kinase Kinase 4/metabolism , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , Substantia Nigra/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/enzymology , Brain Injuries, Traumatic/pathology , Corpus Striatum/enzymology , Corpus Striatum/injuries , Corpus Striatum/pathology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Mice , Neural Pathways/drug effects , Neural Pathways/enzymology , Neural Pathways/injuries , Neural Pathways/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Phosphorylation , Random Allocation , Resveratrol , Substantia Nigra/enzymology , Substantia Nigra/injuries , Substantia Nigra/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Article in English | MEDLINE | ID: mdl-27782041

ABSTRACT

Neonicotinoids are considered safe because of their low affinities to mammalian nicotinic acetylcholine receptors (nAChRs) relative to insect nAChRs. However, because of importance of nAChRs in mammalian brain development, there remains a need to establish the safety of chronic neonicotinoid exposures with regards to children's health. Here we examined the effects of longterm (14 days) and low dose (1 µM) exposure of neuron-enriched cultures from neonatal rat cerebellum to nicotine and two neonicotinoids: acetamiprid and imidacloprid. Immunocytochemistry revealed no differences in the number or morphology of immature neurons or glial cells in any group versus untreated control cultures. However, a slight disturbance in Purkinje cell dendritic arborization was observed in the exposed cultures. Next we performed transcriptome analysis on total RNAs using microarrays, and identified significant differential expression (p < 0.05, q < 0.05, ≥1.5 fold) between control cultures versus nicotine-, acetamiprid-, or imidacloprid-exposed cultures in 34, 48, and 67 genes, respectively. Common to all exposed groups were nine genes essential for neurodevelopment, suggesting that chronic neonicotinoid exposure alters the transcriptome of the developing mammalian brain in a similar way to nicotine exposure. Our results highlight the need for further careful investigations into the effects of neonicotinoids in the developing mammalian brain.


Subject(s)
Cerebellum/drug effects , Imidazoles/toxicity , Insecticides/toxicity , Neurons/drug effects , Nicotine/toxicity , Nitro Compounds/toxicity , Pyridines/toxicity , Transcriptome/drug effects , Animals , Cerebellum/embryology , Gene Expression Regulation, Developmental/drug effects , Neonicotinoids , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics
9.
Brain Res Bull ; 127: 195-201, 2016 10.
Article in English | MEDLINE | ID: mdl-27679398

ABSTRACT

Traumatic brain injury triggers a series of damaged processes, such as neuronal death and apoptosis, inflammation and scar formation, which contribute to evolution of brain injury. The present study investigated the neuroprotective effects of batroxobin, a drug widely used clinically for ischemia, in a nigrostriatal pathway injury model. Mice subjected to the nigrostriatal pathway injury were injected with batroxobin (30 BU/kg) or vehicle immediately after injury. The behavioral studies showed that batroxobin could improve the motor function in injured mice in long term. Batroxobin also reduced neuronal apoptosis and inflammation at the acute stage. Moreover, administration of batroxobin attenuated the scar formation and reduced the lesion size at 4 and 14days after brain injury. These results suggest that batroxobin has beneficial effects on the nigrostriatal pathway injury, indicating a potential clinical application.


Subject(s)
Batroxobin/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/injuries , Neuroprotective Agents/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/injuries , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cicatrix/drug therapy , Cicatrix/immunology , Cicatrix/pathology , Corpus Striatum/immunology , Corpus Striatum/pathology , Disease Models, Animal , Male , Mice , Motor Activity/drug effects , Motor Activity/physiology , Neural Pathways/drug effects , Neural Pathways/immunology , Neural Pathways/injuries , Neural Pathways/pathology , Random Allocation , Substantia Nigra/immunology , Substantia Nigra/pathology
10.
Brain Res ; 1648(Pt A): 90-100, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27402431

ABSTRACT

This study was undertaken to examine the function of extracellular signal-regulated kinase (ERK) signaling pathway on the proliferation and activation of microglia/macrophage and astrocytes after brain injury in mice. The result of Western blot showed that p-ERK was immediately activated after injury (<4h), but the duration was short (<4 days). According to immunofluorescence double staining, it was found that at 4 and 8h after injury, p-ERK was expressed in microglia/macrophages, and that more cells were co-expressed by p-ERK and IBA-1 (microglia/macrophage marker) at 8h; at days 1 and 4, p-ERK was expressed in astrocytes, and more cells were co-expressed by p-ERK and GFAP (astrocyte marker) at day 4. After injury, the mice were injected with U0126 (MAPK/ERK signaling pathway inhibitor) via the femoral vein. Compared with those injected with DMSO, the cell number co-expressed by p-ERK and IBA-1 or GFAP significantly decreased (P<0.05). The increase of microglia/macrophage and astrocyte caused by injury was remitted, and the positive cell number significantly decreased (P<0.05). Western blot showed that the expression quantity of IBA-1 and GFAP significantly decreased (P<0.05). Furthermore, the ERK signaling pathway was involved in the proliferation and activation of the two glial cells types and improved long-term neurobehavioral function after brain injury. Therefore, the exploration of the formation mechanism of glial scar after injury and further research on the therapeutic method of neural regeneration are essential.


Subject(s)
MAP Kinase Signaling System/physiology , Neuroglia/metabolism , Animals , Astrocytes/metabolism , Blotting, Western , Brain Injuries/therapy , Butadienes/pharmacology , Butadienes/therapeutic use , Cell Proliferation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Macrophages/metabolism , Male , Mice , Microglia/metabolism , Neuralgia/drug therapy , Neuroglia/physiology , Nitriles/pharmacology , Nitriles/therapeutic use , Phosphorylation/physiology , Signal Transduction
11.
Drugs Real World Outcomes ; 1(1): 7-19, 2014 Dec.
Article in English | MEDLINE | ID: mdl-27747476

ABSTRACT

BACKGROUND: No data on the long-term 'real-world' use of fluvoxamine for the treatment of social anxiety disorder (SAD) in Japanese patients are currently available. OBJECTIVE: To evaluate the long-term safety and efficacy of fluvoxamine for SAD in the clinical setting. METHODS: Japanese patients with SAD who initiated treatment with fluvoxamine were enrolled in this 53-week post-marketing survey from 407 institutions nationwide. Data including rates of adverse drug reactions (ADRs) and efficacy were collected. Overall improvement was assessed using the Clinical Global Impression for Improvement. SAD symptoms and treatment responses were assessed with the Japanese version of the Liebowitz Social Anxiety Scale. RESULTS: From the 1,974 patients surveyed, 1,790 and 1,504 patients were eligible for analysis of safety and efficacy, respectively. ADRs were reported in 18.2 % of patients, with nausea, somnolence, and constipation the most common. Over 50 % of these ADRs developed in the first 4 weeks of treatment. Serious ADRs were reported in 0.8 % of patients and included six cases of suicide attempt and three cases of suicidal ideation. Response to fluvoxamine was reported in 78.4 % of patients. In patients comorbid with depression, improvement in SAD symptoms with fluvoxamine treatment was significantly affected by clinical improvement in the depression. CONCLUSIONS: These findings support the long-term safety and efficacy of fluvoxamine in patients with SAD. Most ADRs developed during the early treatment phase, and higher doses during the later phase were not associated with an increase in ADRs.

12.
Nat Commun ; 4: 2740, 2013.
Article in English | MEDLINE | ID: mdl-24220492

ABSTRACT

Extracellular factors that inhibit axon growth and intrinsic factors that promote it affect neural regeneration. Therapies targeting any single gene have not yet simultaneously optimized both types of factors. Chondroitin sulphate (CS), a glycosaminoglycan, is the most abundant extracellular inhibitor of axon growth. Here we show that mice carrying a gene knockout for CS N-acetylgalactosaminyltransferase-1 (T1), a key enzyme in CS biosynthesis, recover more completely from spinal cord injury than wild-type mice and even chondroitinase ABC-treated mice. Notably, synthesis of heparan sulphate (HS), a glycosaminoglycan promoting axonal growth, is also upregulated in TI knockout mice because HS-synthesis enzymes are induced in the mutant neurons. Moreover, chondroitinase ABC treatment never induces HS upregulation. Taken together, our results indicate that regulation of a single gene, T1, mediates excellent recovery from spinal cord injury by optimizing counteracting effectors of axon regeneration--an extracellular inhibitor of CS and intrinsic promoters, namely, HS-synthesis enzymes.


Subject(s)
Chondroitin Sulfates/biosynthesis , N-Acetylgalactosaminyltransferases/metabolism , Spinal Cord Injuries/metabolism , Animals , Gene Expression Regulation, Enzymologic , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , Spinal Cord Injuries/genetics
13.
Anat Rec (Hoboken) ; 296(6): 971-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23564738

ABSTRACT

The present study examined gamma-aminobutyric acid B (GABAB ) receptor, GABA, choline acetyltransferase (ChAT), and neuronal nitric oxide synthase (nNOS) immunoreactivities in the mouse adrenal medulla. GABAB receptor immunoreactivity was seen in numerous chromaffin cells and in a few ganglion cells of the adrenal medulla. By using a formaldehyde-induced fluorescence (FIF) method, GABAB receptor immunoreactivity was observed in numerous adrenaline (A) cells, but not in noradrenaline (NA) cells showing blue-white fluorescence. This suggests that GABAB receptors may be present in the A cells and be related to the secretory activity of A cells but not NA cells in the mouse adrenal medulla. GABAB receptor immunoreactive ganglion cells were shown to be nNOS immunopositive by using a double immunostaining method. Weak GABA immunoreactivity was visible in some chromaffin cells and in the numerous nerve fibers of the medulla. By using the FIF method, weak GABA-immunoreactive chromaffin cells were shown to be in the NA cells showing blue-white fluorescence. GABA-immunoreactive nerve fibers were in dense contact in A cells, but not NA cells. GABA-immunoreactive nerve fibers closely contacted a few ganglion cells. Numerous GABA-immunoreactive nerve fibers in the medulla showed ChAT immunoreactive. This result suggests that GABA and acetylcholine may be released from the same nerve fibers and may have a secretory effect on the A cells of the medulla.


Subject(s)
Adrenal Medulla/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Choline O-Acetyltransferase/metabolism , Immunohistochemistry , Male , Mice , Nitric Oxide Synthase Type I/metabolism
14.
J Neurotrauma ; 30(5): 413-25, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23438307

ABSTRACT

Dermatan sulfate (DS) is synthesized from chondroitin sulfate (CS) by epimerization of glucuronic acid of CS to yield iduronic acid. In the present study, the role of CS and DS was examined in mice that received transection of nigrostriatal dopaminergic pathway followed by injection of glycosaminoglycan degrading enzymes into the lesion site. Two weeks after injury, fibrotic and glial scars were formed around the lesion, and transected axons did not regenerate beyond the fibrotic scar. Injection of chondroitinase ABC (ChABC), which degrades both CS and DS, completely suppressed the fibrotic scar formation, reduced the glial scar, and promoted the regeneration of dopaminergic axons. Injection of the DS-degrading enzyme chondroitinase B (ChB) also yielded similar results. By contrast, injection of chondroitinase AC (ChAC), a CS-degrading enzyme, did not suppress the fibrotic and glial scar formation, but reduced CS immunoreactivity and promoted the axonal regeneration. Addition of transforming growth factor-ß1 (TGF-ß1) to a co-culture of meningeal fibroblasts and cerebral astrocytes induces a fibrotic scar-like cell cluster. The effect of TGF-ß1 on cluster formation was suppressed by treatment with ChABC or ChB, but not by ChAC. TGF-ß1-induced cell cluster repelled neurites of neonatal cerebellar neurons, but addition of ChABC or ChAC suppressed the inhibitory property of clusters on neurite outgrowth. The present study is the first to demonstrate that DS and CS play different functions after brain injury: DS is involved in the lesion scar formation, and CS inhibits axonal regeneration.


Subject(s)
Axons/physiology , Brain Injuries/metabolism , Chondroitin Sulfates/metabolism , Cicatrix/metabolism , Dermatan Sulfate/metabolism , Nerve Regeneration/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Axons/pathology , Brain Injuries/pathology , Coculture Techniques , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Male , Mice , Mice, Inbred ICR , Rats , Rats, Sprague-Dawley
15.
Cell Rep ; 3(2): 458-71, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23395638

ABSTRACT

Accumulating evidence suggests that many brain diseases are associated with defects in neuronal migration, suggesting that this step of neurogenesis is critical for brain organization. However, the molecular mechanisms underlying neuronal migration remain largely unknown. Here, we identified the zinc-finger transcriptional repressor RP58 as a key regulator of neuronal migration via multipolar-to-bipolar transition. RP58(-/-) neurons exhibited severe defects in the formation of leading processes and never shifted to the locomotion mode. Cre-mediated deletion of RP58 using in utero electroporation in RP58(flox/flox) mice revealed that RP58 functions in cell-autonomous multipolar-to-bipolar transition, independent of cell-cycle exit. Finally, we found that RP58 represses Ngn2 transcription to regulate the Ngn2-Rnd2 pathway; Ngn2 knockdown rescued migration defects of the RP58(-/-) neurons. Our findings highlight the critical role of RP58 in multipolar-to-bipolar transition via suppression of the Ngn2-Rnd2 pathway in the developing cerebral cortex.


Subject(s)
Cerebral Cortex/growth & development , Neurons/metabolism , Repressor Proteins/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Movement , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Development , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Neurons/cytology , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics
16.
J Neurosci Res ; 91(3): 335-48, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239230

ABSTRACT

We recently reported that a new monoclonal antibody, 4F2, which labels oligodendroglial lineage cells, recognizes a DEAD-box RNA helicase Ddx54 and that Ddx54 binds to myelin basic protein (MBP) in brain and cultured oligodendrocytes. To elucidate the biological function of Ddx54, we generated a recombinant adenovirus, Ad-shRNA:Ddx54, expressing a short hairpin RNA to silence endogenous Ddx54 protein. The virus was intraventricularly injected into the brains of mice on postnatal day (PD) 2. The brains at PD 9 were then analyzed by immunohistochemistry. In untreated normal brain sections, as well as control brains that had been injected with Ad-ß-Gal, myelination of axons occurred in the corpus callosum with filamentous patterns of immunosignals of myelin-associated glycoprotein (MAG) and MBP. In Ad-shRNA:Ddx54-injected brain, substantial amounts of MAG and MBP immunosignals were present, but MBP immunosignals accumulated in the subplate layer and did not intrude into the emerging white matter. Immunoblot analysis revealed that Ddx54 knockdown caused a significant decrease in the level of 21.5 kDa MBP isoform and Ddx54, but the amount of Olig2; 2',3'-cyclic nucleotide 3' phosphodiesterase; MAG; three MBP isoforms (14, 17.5, and 18 kDa); and QKI-5, QKI-6, and QKI-7 proteins remained unchanged. Transfection of the Ddx54 expression vector into luciferase reporter-introduced neuroepithelial cells resulted in upregulated MBP promoter activity. Immunoprecipitation of Ddx54 protein in MBP-transfected HEK293 cells indicated that Ddx54 may directly interact with MBP mRNA. These results suggest that Ddx54 protein play an important role in central nervous system myelination, presumably in myelin sheath formation after the differentiation of oligodendrocytes.


Subject(s)
Brain/cytology , Brain/physiology , DEAD-box RNA Helicases/physiology , Myelin Sheath/physiology , Neoplasm Proteins/physiology , Oligodendroglia/physiology , Animals , Animals, Newborn , Brain/metabolism , Cell Differentiation/physiology , Cells, Cultured , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Pregnancy
17.
Nutrients ; 4(10): 1504-17, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-23201768

ABSTRACT

The aim of this study was to evaluate the effects of egg white protein compared to carbohydrate intake prior to exercise on fat free mass (FFM), one repetition maximum (1RM) muscle strength and blood biochemistry in female athletes. Thirty healthy female collegiate athletes were recruited for this study and matched by sport type, body fat percentage and 1RM leg curl muscle strength. Participants were randomly divided into two groups: protein group (15.0 g egg white protein; 75 kcal) and carbohydrate group (17.5 g maltodextrin, 78 kcal). Supplements were administered daily at the same time in a double-blind manner prior to training during an 8-week period. Measurements were performed before and after the 8-week regimen. The mean dietary energy intake did not change throughout the study period. FFM and 1RM assessments (i.e., leg curl, leg extension, squat, and bench press) increased in both groups. Furthermore, serum urea and serum citrulline levels after the 8-week regimen increased significantly only in the protein group. Our findings indicated that compared to the carbohydrate supplement, the protein supplement was associated with some changes in protein metabolites but not with changes in body composition or muscle strength.


Subject(s)
Amino Acids/blood , Dietary Carbohydrates/pharmacology , Dietary Proteins/pharmacology , Egg Proteins/pharmacology , Egg White/chemistry , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Adolescent , Adult , Athletes , Citrulline/blood , Dietary Supplements , Double-Blind Method , Female , Humans , Leg , Muscle, Skeletal/physiology , Resistance Training , Urea/blood , Young Adult
18.
J Neurosci ; 32(19): 6587-99, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22573681

ABSTRACT

Axonal outgrowth is a coordinated process of cytoskeletal dynamics and membrane trafficking; however, little is known about proteins responsible for regulating the membrane supply. LMTK1 (lemur kinase 1)/AATYK1 (apoptosis-associated tyrosine kinase 1) is a serine/threonine kinase that is highly expressed in neurons. We recently reported that LMTK1 plays a role in recycling endosomal trafficking in CHO-K1 cells. Here we explore the role of LMTK1 in axonal outgrowth and its regulation by Cdk5 using mouse brain cortical neurons. LMTK1 was expressed and was phosphorylated at Ser34, the Cdk5 phosphorylation site, at the time of axonal outgrowth in culture and colocalized with Rab11A, the small GTPase that regulates recycling endosome traffic, at the perinuclear region and in the axon. Overexpression of the unphosphorylated mutant LMTK1-S34A dramatically promoted axonal outgrowth in cultured neurons. Enhanced axonal outgrowth was diminished by the inactivation of Rab11A, placing LMTK1 upstream of Rab11A. Unexpectedly, the downregulation of LMTK1 by knockdown or gene targeting also significantly enhanced axonal elongation. Rab11A-positive vesicles were transported anterogradely more quickly in the axons of LMTK1-deficient neurons than in those of wild-type neurons. The enhanced axonal outgrowth was reversed by LMTK1-WT or the LMTK1-S34D mutant, which mimics the phosphorylated state, but not by LMTK1-S34A. Thus, LMTK1 can negatively control axonal outgrowth by regulating Rab11A activity in a Cdk5-dependent manner, and Cdk5-LMTK1-Rab11 is a novel signaling pathway involved in axonal outgrowth.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Axons/physiology , Cyclin-Dependent Kinase 5/physiology , Growth Cones/physiology , Protein-Tyrosine Kinases/physiology , rab GTP-Binding Proteins/physiology , Animals , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Axons/enzymology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Growth Cones/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Phosphorylation/physiology , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , rab GTP-Binding Proteins/antagonists & inhibitors
19.
PLoS One ; 7(2): e32432, 2012.
Article in English | MEDLINE | ID: mdl-22393406

ABSTRACT

BACKGROUND: Acetamiprid (ACE) and imidacloprid (IMI) belong to a new, widely used class of pesticide, the neonicotinoids. With similar chemical structures to nicotine, neonicotinoids also share agonist activity at nicotinic acetylcholine receptors (nAChRs). Although their toxicities against insects are well established, their precise effects on mammalian nAChRs remain to be elucidated. Because of the importance of nAChRs for mammalian brain function, especially brain development, detailed investigation of the neonicotinoids is needed to protect the health of human children. We aimed to determine the effects of neonicotinoids on the nAChRs of developing mammalian neurons and compare their effects with nicotine, a neurotoxin of brain development. METHODOLOGY/PRINCIPAL FINDINGS: Primary cultures of cerebellar neurons from neonatal rats allow for examinations of the developmental neurotoxicity of chemicals because the various stages of neurodevelopment-including proliferation, migration, differentiation, and morphological and functional maturation-can be observed in vitro. Using these cultures, an excitatory Ca(2+)-influx assay was employed as an indicator of neural physiological activity. Significant excitatory Ca(2+) influxes were evoked by ACE, IMI, and nicotine at concentrations greater than 1 µM in small neurons in cerebellar cultures that expressed the mRNA of the α3, α4, and α7 nAChR subunits. The firing patterns, proportion of excited neurons, and peak excitatory Ca(2+) influxes induced by ACE and IMI showed differences from those induced by nicotine. However, ACE and IMI had greater effects on mammalian neurons than those previously reported in binding assay studies. Furthermore, the effects of the neonicotinoids were significantly inhibited by the nAChR antagonists mecamylamine, α-bungarotoxin, and dihydro-ß-erythroidine. CONCLUSIONS/SIGNIFICANCE: This study is the first to show that ACE, IMI, and nicotine exert similar excitatory effects on mammalian nAChRs at concentrations greater than 1 µM. Therefore, the neonicotinoids may adversely affect human health, especially the developing brain.


Subject(s)
Cerebellum/cytology , Imidazoles/pharmacology , Insecticides/pharmacology , Neurons/metabolism , Nicotine/pharmacology , Nitro Compounds/pharmacology , Pyridines/pharmacology , Animals , Animals, Newborn , Brain/metabolism , Bungarotoxins/pharmacology , Calcium/metabolism , Cerebellum/metabolism , Dihydro-beta-Erythroidine/pharmacology , Humans , Mecamylamine/pharmacology , Neonicotinoids , Neurotoxins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
20.
Cell Tissue Res ; 349(1): 169-80, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22362507

ABSTRACT

Traumatic damage to the central nervous system (CNS) destroys the blood-brain barrier (BBB) and provokes the invasion of hematogenous cells into the neural tissue. Invading leukocytes, macrophages and lymphocytes secrete various cytokines that induce an inflammatory reaction in the injured CNS and result in local neural degeneration, formation of a cystic cavity and activation of glial cells around the lesion site. As a consequence of these processes, two types of scarring tissue are formed in the lesion site. One is a glial scar that consists in reactive astrocytes, reactive microglia and glial precursor cells. The other is a fibrotic scar formed by fibroblasts, which have invaded the lesion site from adjacent meningeal and perivascular cells. At the interface, the reactive astrocytes and the fibroblasts interact to form an organized tissue, the glia limitans. The astrocytic reaction has a protective role by reconstituting the BBB, preventing neuronal degeneration and limiting the spread of damage. While much attention has been paid to the inhibitory effects of the astrocytic component of the scars on axon regeneration, this review will cover a number of recent studies in which manipulations of the fibroblastic component of the scar by reagents, such as blockers of collagen synthesis have been found to be beneficial for axon regeneration. To what extent these changes in the fibroblasts act via subsequent downstream actions on the astrocytes remains for future investigation.


Subject(s)
Central Nervous System/pathology , Cicatrix/pathology , Wound Healing , Animals , Axons/pathology , Humans , Nerve Regeneration , Neuroglia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...