Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Annu Rev Pharmacol Toxicol ; 64: 417-433, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-37708431

ABSTRACT

Particles and crystals constitute a unique class of toxic agents that humans are constantly exposed to both endogenously and from the environment. Deposition of particulates in the body is associated with a range of diseases and toxicity. The mechanism by which particulates cause disease remains poorly understood due to the lack of mechanistic insights into particle-biological interactions. Recent research has revealed that many particles and crystals activate the NLRP3 inflammasome, an intracellular pattern-recognition receptor. Activated NLRP3 forms a supramolecular complex with an adaptor protein to activate caspase 1, which in turn activates IL-1ß and IL-18 to instigate inflammation. Genetic ablation and pharmacological inhibition of the NLRP3 inflammasome dampen inflammatory responses to particulates. Nonetheless, how particulates activate NLRP3 remains a challenging question. From this perspective, we discuss our current understanding of and progress on revealing the function and mode of action of the NLRP3 inflammasome in mediating adaptive and pathologic responses to particulates in health and disease.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Caspase 1/metabolism
2.
Nanotoxicology ; 17(3): 249-269, 2023 04.
Article in English | MEDLINE | ID: mdl-37115655

ABSTRACT

Fibrogenic carbon nanotubes (CNTs) induce the polarization of M1 and M2 macrophages in mouse lungs. Polarization of the macrophages regulates the production of proinflammatory and pro-resolving lipid mediators (LMs) to mediate acute inflammation and its resolution in a time-dependent manner. Here we examined the molecular mechanism by which multi-walled CNTs (MWCNTs, Mitsui-7) induce M1 polarization in vitro. Treatment of murine macrophages (J774A.1) with Mitsui-7 MWCNTs increased the expression of Alox5 mRNA and protein in a concentration- and time-dependent manner. The MWCNTs induced the expression of CD68 and that induction persisted for up to 3 days post-exposure. The expression and activity of inducible nitric oxide synthase, an intracellular marker of M1, were increased by MWCNTs. Consistent with M1 polarization, the MWCNTs induced the production and secretion of proinflammatory cytokines tumor necrosis factor-α and interleukin-1ß, and proinflammatory LMs leukotriene B4 (LTB4) and prostaglandin E2 (PGE2). The cell-free media from MWCNT-polarized macrophages induced the migration of neutrophilic cells (differentiated from HL-60), which was blocked by Acebilustat, a specific leukotriene A4 hydrolase inhibitor, or LY239111, an LTB4 receptor antagonist, but not NS-398, a cyclooxygenase 2 inhibitor, revealing LTB4 as a major mediator of neutrophil chemotaxis from MWCNT-polarized macrophages. Knockdown of Alox5 using specific small hairpin-RNA suppressed MWCNT-induced M1 polarization, LTB4 secretion, and migration of neutrophils. Taken together, these findings demonstrate the polarization of M1 macrophages by Mitsui-7 MWCNTs in vitro and that induction of Alox5 is an important mechanism by which the MWCNTs promote proinflammatory responses by boosting M1 polarization and production of proinflammatory LMs.


Subject(s)
Arachidonate 5-Lipoxygenase , Macrophages , Nanotubes, Carbon , Animals , Mice , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Cytokines/metabolism , Leukotriene B4/metabolism , Nanotubes, Carbon/toxicity , Macrophage Activation
3.
Front Immunol ; 11: 1186, 2020.
Article in English | MEDLINE | ID: mdl-32595644

ABSTRACT

Pulmonary exposure to certain engineered nanomaterials (ENMs) causes chronic lesions like fibrosis and cancer in animal models as a result of unresolved inflammation. Resolution of inflammation involves the time-dependent biosynthesis of lipid mediators (LMs)-in particular, specialized pro-resolving mediators (SPMs). To understand how ENM-induced pulmonary inflammation is resolved, we analyzed the inflammatory and pro-resolving responses to fibrogenic multi-walled carbon nanotubes (MWCNTs, Mitsui-7) and low-toxicity fullerenes (fullerene C60, C60F). Pharyngeal aspiration of MWCNTs at 40 µg/mouse or C60F at a dose above 640 µg/mouse elicited pulmonary effects in B6C3F1 mice. Both ENMs stimulated acute inflammation, predominated by neutrophils, in the lung at day 1, which transitioned to histiocytic inflammation by day 7. By day 28, the lesion in MWCNT-exposed mice progressed to fibrotic granulomas, whereas it remained as alveolar histiocytosis in C60F-exposed mice. Flow cytometric profiling of whole lung lavage (WLL) cells revealed that neutrophil recruitment was the greatest at day 1 and declined to 36.6% of that level in MWCNT- and 16.8% in C60F-treated mice by day 7, and to basal levels by day 28, suggesting a rapid initiation phase and an extended resolution phase. Both ENMs induced high levels of proinflammatory leukotriene B4 (LTB4) and prostaglandin E2 (PGE2) with peaks at day 1, and high levels of SPMs resolvin D1 (RvD1) and E1 (RvE1) with peaks at day 7. MWCNTs and C60F induced time-dependent polarization of M1 macrophages with a peak at day 1 and subsequently of M2 macrophages with a peak at day 7 in the lung, accompanied by elevated levels of type 1 or type 2 cytokines, respectively. M1 macrophages exhibited preferential induction of arachidonate 5-lipoxygenase activating protein (ALOX5AP), whereas M2 macrophages had a high level expression of arachidonate 15-lipoxygenase (ALOX15). Polarization of macrophages in vitro differentially induced ALOX5AP in M1 macrophages or ALOX15 in M2 macrophages resulting in increased preferential biosynthesis of proinflammatory LMs or SPMs. MWCNTs increased the M1- or M2-specific production of LMs accordingly. These findings support a mechanism by which persistent ENM-induced neutrophilic inflammation is actively resolved through time-dependent polarization of macrophages and enhanced biosynthesis of specialized LMs via distinct ALOX pathways.


Subject(s)
Fullerenes/toxicity , Macrophage Activation/immunology , Macrophages/immunology , Nanotubes, Carbon/toxicity , Pneumonia/chemically induced , Pneumonia/immunology , Animals , Macrophages/drug effects , Mice , Pneumonia/pathology
4.
J Breast Cancer ; 21(2): 182-189, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29963114

ABSTRACT

PURPOSE: There are few reports from Asian countries about the long-term results of aromatase inhibitor adjuvant treatment for breast cancer. This observational study aimed to evaluate the long-term effects of letrozole in postmenopausal Korean women with operable breast cancer. METHODS: Self-reported quality of life (QoL) scores were serially assessed for 3 years during adjuvant letrozole treatment using the Korean version of the Functional Assessment of Cancer Therapy-Breast questionnaires (version 3). Changes in bone mineral density (BMD) and serum cholesterol levels were also examined. RESULTS: All 897 patients received the documented informed consent form and completed a baseline questionnaire before treatment. Adjuvant chemotherapy was administered to 684 (76.3%) subjects, and 410 (45.7%) and 396 (44.1%) patients had stage I and II breast cancer, respectively. Each patient completed questionnaires at 3, 6, 12, 18, 24, 30, and 36 months after enrollment. Of 897 patients, 749 (83.5%) completed the study. The dropout rate was 16.5%. The serial trial outcome index, the sum of the physical and functional well-being subscales, increased gradually and significantly from baseline during letrozole treatment (p<0.001). The mean serum cholesterol level increased significantly from 199 to 205 after 36 months (p=0.042). The mean BMD significantly decreased from -0.39 at baseline to -0.87 after 36 months (p<0.001). CONCLUSION: QoL gradually improved during letrozole treatment. BMD and serum cholesterol level changes were similar to those in Western countries, indicating that adjuvant letrozole treatment is well tolerated in Korean women, with minimal ethnic variation.

5.
Biosci Rep ; 38(1)2018 02 28.
Article in English | MEDLINE | ID: mdl-29330223

ABSTRACT

Activity of neprilysin (NEP), the major protease which cleaves amyloid-ß peptide (Aß), is reportedly reduced in the brains of patients with Alzheimer's disease (AD). Accumulation of Aß generates reactive oxygen species (ROS) such as 4-hydroxynonenal (HNE), and then reduces activities of Aß-degrading enzymes including NEP. Xanthorrhizol (Xan), a natural sesquiterpenoid, has been reported to possess antioxidant and anti-inflammatory properties. The present study examined the effects of Xan on HNE- or oligomeric Aß42-induced oxidative modification of NEP protein. Xan was added to the HNE- or oligomeric Aß42-treated SK-N-SH human neuroblastoma cells and then levels, oxidative modification and enzymatic activities of NEP protein were measured. Increased HNE levels on NEP proteins and reduced enzymatic activities of NEP were observed in the HNE- or oligomeric Aß42-treated cells. Xan reduced HNE levels on NEP proteins and preserved enzymatic activities of NEP in HNE- or oligomeric Aß42-treated cells. Xan reduced Aß42 accumulation and protected neurones against oligomeric Aß42-induced neurotoxicity through preservation of NEP activities. These findings indicate that Xan possesses therapeutic potential for the treatment of neurodegenerative diseases, including AD, and suggest a potential mechanism for the neuroprotective effects of antioxidants for the prevention of AD.


Subject(s)
Alzheimer Disease/drug therapy , Antioxidants/administration & dosage , Neprilysin/metabolism , Phenols/administration & dosage , Aldehydes/administration & dosage , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line , Humans , Neprilysin/antagonists & inhibitors , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Neuroprotective Agents/administration & dosage , Oxidation-Reduction , Oxidative Stress/drug effects , Peptide Fragments/chemistry , Peptide Fragments/genetics , Reactive Oxygen Species/metabolism
6.
J Alzheimers Dis ; 58(2): 521-535, 2017.
Article in English | MEDLINE | ID: mdl-28482641

ABSTRACT

Bryostatin 1, a potent activator of protein kinase C epsilon (PKCɛ), has been shown to reverse synaptic loss and facilitate synaptic maturation in animal models of Alzheimer's disease (AD), Fragile X, stroke, and other neurological disorders. In a single-dose (25 µg/m2) randomized double-blind Phase IIa clinical trial, bryostatin levels reached a maximum at 1-2 h after the start of infusion. In close parallel with peak blood levels of bryostatin, an increase of PBMC PKCɛ was measured (p = 0.0185) within 1 h from the onset of infusion. Of 9 patients with a clinical diagnosis of AD, of which 6 received drug and 3 received vehicle within a double-blind protocol, bryostatin increased the Mini-Mental State Examination (MMSE) score by +1.83±0.70 unit at 3 h versus -1.00±1.53 unit for placebo. Bryostatin was well tolerated in these AD patients and no drug-related adverse events were reported. The 25 µg/m2 administered dose was based on prior clinical experience with three Expanded Access advanced AD patients treated with bryostatin, in which return of major functions such as swallowing, vocalization, and word recognition were noted. In one Expanded Access patient trial, elevated PKCɛ levels closely tracked cognitive benefits in the first 24 weeks as measured by MMSE and ADCS-ADL psychometrics. Pre-clinical mouse studies showed effective activation of PKCɛ and increased levels of BDNF and PSD-95. Together, these Phase IIa, Expanded Access, and pre-clinical results provide initial encouragement for bryostatin 1 as a potential treatment for AD.


Subject(s)
Alzheimer Disease , Antipsychotic Agents/therapeutic use , Bryostatins/therapeutic use , Cognition Disorders , Protein Kinase C-epsilon/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/etiology , Disks Large Homolog 4 Protein/metabolism , Double-Blind Method , Female , Humans , Male , Mental Status Schedule , Mice , Mice, Inbred C57BL , Middle Aged , Neuropsychological Tests , Phosphopyruvate Hydratase/metabolism , Psychometrics , Synaptophysin/metabolism , Time Factors
7.
J Biol Chem ; 292(15): 6402-6413, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28264928

ABSTRACT

An improved understanding of the molecular mechanisms in synapse formation provides insight into both learning and memory and the etiology of neurodegenerative disorders. Coactivator-associated arginine methyltransferase 1 (CARM1) is a protein methyltransferase that negatively regulates synaptic gene expression and inhibits neuronal differentiation. Despite its regulatory function in neurons, little is known about the CARM1 cellular location and its role in dendritic maturation and synapse formation. Here, we examined the effects of CARM1 inhibition on dendritic spine and synapse morphology in the rat hippocampus. CARM1 was localized in hippocampal post-synapses, with immunocytochemistry and electron microscopy revealing co-localization of CARM1 with post-synaptic density (PSD)-95 protein, a post-synaptic marker. Specific siRNA-mediated suppression of CARM1 expression resulted in precocious dendritic maturation, with increased spine width and density at sites along dendrites and induction of mushroom-type spines. These changes were accompanied by a striking increase in the cluster size and number of key synaptic proteins, including N-methyl-d-aspartate receptor subunit 2B (NR2B) and PSD-95. Similarly, pharmacological inhibition of CARM1 activity with the CARM1-specific inhibitor AMI-1 significantly increased spine width and mushroom-type spines and also increased the cluster size and number of NR2B and cluster size of PSD-95. These results suggest that CARM1 is a post-synaptic protein that plays roles in dendritic maturation and synaptic formation and that spatiotemporal regulation of CARM1 activity modulates neuronal connectivity and improves synaptic dysfunction.


Subject(s)
Dendrites/enzymology , Hippocampus/enzymology , Post-Synaptic Density/enzymology , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cells, Cultured , Disks Large Homolog 4 Protein , Hippocampus/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/cytology , Spinal Cord/enzymology
8.
J Alzheimers Dis ; 43(2): 491-509, 2015.
Article in English | MEDLINE | ID: mdl-25125477

ABSTRACT

In Alzheimer's disease (AD) transgenic mice, activation of synaptogenic protein kinase C ε (PKCε) was found to prevent synaptotoxic amyloid-ß (Aß)-oligomer elevation, PKCε deficits, early synaptic loss, cognitive deficits, and amyloid plaque formation. In humans, to study the role of PKCε in the pathophysiology of AD and to evaluate its possible use as an early AD-biomarker, we examined PKCε and Aß in the brains of autopsy-confirmed AD patients (n = 20) and age-matched controls (AC, n = 19), and in skin fibroblast samples from AD (n = 14), non-AD dementia patients (n = 14), and AC (n = 22). Intraneuronal Aß levels were measured immunohistochemically (using an Aß-specific antibody) in hippocampal pyramidal cells of human autopsy brains. PKCε was significantly lower in the hippocampus and temporal pole areas of AD brains, whereas Aß levels were significantly higher. The ratio of PKCε to Aß in individual CA1 pyramidal cells was markedly lower in the autopsy AD brains versus controls. PKCε was inversely correlated with Aß levels in controls, whereas in AD patients, PKCε showed no significant correlation with Aß. In autopsy brains, PKCε decreased as the Braak score increased. Skin fibroblast samples from AD patients also demonstrated a deficit in PKCε compared to controls and an AD-specific change in the Aß-oligomer effects on PKCε. Together, these data demonstrate that the relationship between Aß levels and PKCε is markedly altered in AD patients' brains and skin fibroblasts, reflecting a loss of protective effect of PKCε against toxic Aß accumulation. These changes of PKCε levels in human skin fibroblasts may provide an accurate, non-invasive peripheral AD biomarker.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Brain/enzymology , Fibroblasts/enzymology , Protein Kinase C/deficiency , Skin/pathology , Adult , Aged , Aged, 80 and over , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Analysis of Variance , Autopsy , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Humans , Male , Middle Aged , Protein Kinase C/genetics , RNA, Messenger/metabolism
9.
PLoS One ; 9(5): e97756, 2014.
Article in English | MEDLINE | ID: mdl-24848988

ABSTRACT

Amyloid-beta (Aß) peptide accumulation in the brain is a pathological hallmark of all forms of Alzheimer's disease. An imbalance between Aß production and clearance from the brain may contribute to accumulation of neurotoxic Aß and subsequent synaptic loss, which is the strongest correlate of the extent of memory loss in AD. The activity of neprilysin (NEP), a potent Aß-degrading enzyme, is decreased in the AD brain. Expression of HuD, an mRNA-binding protein important for synaptogenesis and neuronal plasticity, is also decreased in the AD brain. HuD is regulated by protein kinase Cε (PKCε), and we previously demonstrated that PKCε activation decreases Aß levels. We hypothesized that PKCε acts through HuD to stabilize NEP mRNA, modulate its localization, and support NEP activity. Conversely, loss of PKCε-activated HuD in AD leads to decreased NEP activity and accumulation of Aß. Here we show that HuD is associated with NEP mRNA in cultures of human SK-N-SH cells. Treatment with bryostatin, a PKCε-selective activator, enhanced NEP association with HuD and increased NEP mRNA stability. Activation of PKCε also increased NEP protein levels, increased NEP phosphorylation, and induced cell surface expression. In addition, specific PKCε activation directly stimulated NEP activity, leading to degradation of a monomeric form of Aß peptide and decreased Aß neuronal toxicity, as measured by cell viability. Bryostatin treatment also rescued Aß-mediated inhibition of HuD-NEP mRNA binding, NEP protein expression, and NEP cell membrane translocation. These results suggest that PKCε activation reduces Aß by up-regulating, via the mRNA-binding protein HuD, Aß-degrading enzymes such as NEP. Thus, PKCε activation may have therapeutic efficacy for AD by reducing neurotoxic Aß accumulation as well as having direct anti-apoptotic and synaptogenic effects.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/cytology , ELAV Proteins/metabolism , Neprilysin/genetics , Neurons/metabolism , Protein Kinase C-epsilon/metabolism , Proteolysis , Amyloid beta-Peptides/toxicity , Cell Line, Tumor , Cell Membrane/metabolism , Enzyme Activation , Humans , Neurons/cytology , Phosphorylation , Protein Transport , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/genetics
10.
J Pharmacol Exp Ther ; 349(3): 393-401, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659806

ABSTRACT

Fragile X syndrome (FXS) is caused by transcriptional silencing in neurons of the FMR1 gene product, fragile X mental retardation protein (FMRP), a repressor of dendritic mRNA translation. The lack of FMRP leads to dysregulation of synaptically driven protein synthesis and impairments of intellect, cognition, and behavior, a disorder that currently has no effective therapeutics. Fragile X mice were treated with chronic bryostatin-1, a relatively selective protein kinase ε activator with pharmacological profiles of rapid mGluR desensitization, synaptogenesis, and synaptic maturation/repairing. Differences in the major FXS phenotypes, synapses, and cognitive functions were evaluated and compared among the age-matched groups. Long-term treatment with bryostatin-1 rescues adult fragile X mice from the disorder phenotypes, including normalization of most FXS abnormalities in hippocampal brain-derived neurotrophic factor expression and secretion, postsynaptic density-95 levels, glycogen synthase kinase-3ß phosphorylation, transformation of immature dendritic spines to mature synapses, densities of the presynaptic and postsynaptic membranes, and spatial learning and memory. Our results show that synaptic and cognitive function of adult FXS mice can be normalized through pharmacologic treatment and that bryostatin-1-like agents may represent a novel class of drugs to treat fragile X mental retardation even after postpartum brain development has largely completed.


Subject(s)
Bryostatins/therapeutic use , Fragile X Syndrome/drug therapy , Hippocampus/drug effects , Maze Learning/drug effects , Memory/drug effects , Spatial Behavior/drug effects , Synapses/drug effects , Animals , Bryostatins/administration & dosage , Dendritic Spines/drug effects , Dendritic Spines/pathology , Disease Models, Animal , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Electron , Protein Kinase C-epsilon/metabolism , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure
11.
Biochem Biophys Res Commun ; 442(3-4): 165-70, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24269817

ABSTRACT

Autophagy is a cellular degradation process for cellular aggregates and unneeded cellular compartments including damaged mitochondria, ER, and peroxisomes. Melanosome is cellular organelle that is the cellular site of generation, storage and transports of melanin in melanocytes. Despite potential importance of autophagy, the role of autophagy in melanogenesis and melanosome autophagy are largely unknown. In here, we identified 3'-hydroxydaidzein (3'-ODI) as an autophagy inducer from a phytochemical library screening. Treatment with 3'-ODI significantly reduced α-MSH-mediated melanogenesis but efficiently increased autophagy both in melanoma cells and melanocytes. Furthermore, inhibition of autophagy significantly reduced the anti-melanogenic effects of 3'-ODI in α-MSH-stimulated melanoma cells. Taken together, these results suggest that autophagy mediates anti-melanogenic activity of 3'-ODI.


Subject(s)
Autophagy/drug effects , Isoflavones/pharmacology , Melanins/antagonists & inhibitors , Melanocytes/drug effects , Melanosomes/drug effects , Animals , Autophagy/genetics , Autophagy-Related Protein 5 , Cell Line, Tumor , Melanins/biosynthesis , Melanocytes/metabolism , Melanosomes/metabolism , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , RNA Interference , alpha-MSH/pharmacology
12.
Hippocampus ; 22(12): 2303-19, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22736542

ABSTRACT

HuD protein is an RNA-binding protein involved in post-transcriptional regulation of gene expression for synaptogenesis, neuronal differentiation, and learning and memory, and is up-regulated and redistributed by a protein kinase C (PKC)-dependent pathway in neurons. Here, we show a PKC-regulated mechanism on HuD-mediated mRNA stability and expression of several neurotrophic factors (NTFs) in cultured hippocampal neurons. HuD pull-down assays showed that HuD is associated with brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and neurotrophin (NT)-3 mRNAs. Reduction of HuD expression with short hairpin RNAs decreased BDNF, NGF, and NT-3 mRNAs and NTFs expression. Bryostatin, a PKC activator, treatment enhanced their association with HuD and increased these transcripts' stability. Bryostatin induced HuD phosphorylation, which was inhibited by Ro 32-0432, a specific PKC inhibitor. Activated PKC specifically phosphorylated coactivator-associated arginine methyltransferase 1 (CARM1), which methylates HuD and negatively modulates HuD-mRNA interactions during neuronal differentiation, and inhibited its methyltransferase activity, resulting in decrease in CARM1-mediated HuD methylation. Furthermore cotreatment of bryostatin and AMI-1, a specific CARM1 inhibitor, potentiated PKC-dependent HuD-mRNA interactions and enhanced dendritic arborization. These results demonstrate that PKC may play an important role in neuronal differentiation and synaptogenesis via stimulating HuD-mediated mRNA stability and inhibiting CARM1 in hippocampal neurons.


Subject(s)
ELAV Proteins/metabolism , Nerve Growth Factors/biosynthesis , Neurogenesis/physiology , Protein Kinase C/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cell Differentiation/physiology , Chromatography, High Pressure Liquid , Dendrites/metabolism , Dendrites/ultrastructure , ELAV-Like Protein 4 , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Hippocampus/metabolism , Hippocampus/ultrastructure , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Neurons/metabolism , Neurons/ultrastructure , RNA Stability , RNA, Messenger , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
13.
Hippocampus ; 22(3): 455-65, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21254302

ABSTRACT

Removal of the cholinergic innervation to the hippocampus via selective immunolesions of septohippocampal cholinergic neurons induces dysfunction of the hypothalamic-pituitary-adrenocortical (HPA) axis and decreases glucocorticoid receptor (GR) mRNA. This study examined whether removal of the cholinergic innervation decreased GR protein levels and induced changes in the interaction between GR and the cytoplasmic catalytic subunit of protein kinase A (PKAc) in the hippocampus. In lesioned animals, GR protein levels were markedly decreased in the nucleus, but not in the cytosol of hippocampal neurons, whereas mineralocorticoid receptor (MR) levels remained unchanged in both the nucleus and cytosol. PKAc levels did not differ between lesioned and control groups, but PKAc activity was reduced in lesion tissue compared with the controls. The interaction between GR and PKAc was also decreased in the hippocampus without cholinergic input. These results indicate that degeneration of septohippocampal cholinergic neurons leads to reduced PKAc activity in the hippocampus which, in turn, alters GR signaling. The altered GR signaling induced by the degeneration of basal forebrain cholinergic neurons may contribute to dysfunction of the HPA axis in aged animals and patients with Alzheimer's disease (AD) and lead to neuropsychiatric symptoms that occur throughout the course of AD.


Subject(s)
Cholinergic Neurons/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Hippocampus/metabolism , Prosencephalon/metabolism , Receptors, Glucocorticoid/metabolism , Signal Transduction/physiology , Animals , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Male , Neural Pathways/metabolism , Pituitary-Adrenal System/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/metabolism
14.
Cell Signal ; 20(5): 825-35, 2008 May.
Article in English | MEDLINE | ID: mdl-18262389

ABSTRACT

During central nervous system development, growth factors and their associated receptor protein tyrosine kinases regulate many neuronal functions such as neurite extension and dendrite maturation. Hepatocyte growth factor (HGF) and its receptor, c-Met, can promote formation of neurites and enhance elaboration of dendrites in mature neurons, but their effects on the early stages of dendrite maturation in hippocampal neurons and the signaling pathways by which they promote dendrite formation have not been studied. Exogenous HGF treatment effectively enhanced the phosphorylation and activation of c-Met in cultured hippocampal neurons at 4 days in vitro. HGF treatment increased the number of dendrites and promoted dendrite elongation in these neurons. Consistent with these results, HGF activated Akt, which phosphorylates glycogen synthase kinase-3beta (GSK-3beta) to inactivate it, and reduced phosphorylation of microtubule-associated protein 2 (MAP2), which can promote microtubule polymerization and dendrite elongation when dephosphorylated. Conversely, pharmacological inhibition of c-Met with its specific inhibitor, PHA-665752, or genetic knock-down of c-Met with short hairpin RNAs (shRNAs) suppressed HGF-induced phosphorylation of Akt and GSK-3beta, increased phosphorylation of MAP2, and reduced dendrite number and length in cultured hippocampal neurons. Moreover, suppressing c-Met with PHA-665752 or by shRNA decreased MAP2 expression. Inhibiting Akt activity with the phosphoinositide-3-kinase inhibitor LY294002 or Akt inhibitor X suppressed HGF-induced phosphorylation of GSK-3beta, increased MAP2 phosphorylation, and blocked the ability of HGF to enhance dendritic length. These observations indicate that HGF and c-Met can regulate the early stages of dendrite maturation via activation of the Akt/GSK-3beta pathway.


Subject(s)
Hepatocyte Growth Factor/pharmacology , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-met/metabolism , Animals , Base Sequence , Cell Differentiation/drug effects , Cells, Cultured , DNA Primers/genetics , Dendrites/drug effects , Dendrites/ultrastructure , Gene Expression Regulation, Developmental , Hepatocyte Growth Factor/genetics , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Indoles/pharmacology , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Rats , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfones/pharmacology
15.
Biol Pharm Bull ; 29(6): 1212-6, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16755019

ABSTRACT

The antioxidant and anti-inflammatory properties of the marine red alga Neorhodomela aculeate (N. aculeata) Masuda were investigated with neuronal and microglial cells. Extracts of N. aculeata had potent neuroprotective effects on glutamate-induced neurotoxicity and inhibited reactive oxygen species (ROS) generation in the murine hippocampal HT22 cell line. Also, extracts of N. aculeata inhibited H2O2-induced lipid peroxidation in rat brain homogenates. The properties of the extract as an anti-inflammatory agent were investigated in microglial activation by interferon-gamma (IFN-gamma): it reduced the inducible nitric oxide synthase that consequently resulted in the reduction of nitric oxide. These results suggest that the marine red alga N. aculeata could be considered as a potential source for reducing reactive oxygen species and inflammation related to neurological diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Hippocampus/drug effects , Microglia/drug effects , Rhodophyta/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Antioxidants/isolation & purification , Biphenyl Compounds/chemistry , Cell Line , Cell Survival/drug effects , Hippocampus/cytology , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Lipid Peroxidation/drug effects , Methanol/chemistry , Mice , Microglia/cytology , Microglia/metabolism , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacology , Nitric Oxide/antagonists & inhibitors , Picrates/chemistry , Rats , Reactive Oxygen Species/metabolism
16.
Neurosci Lett ; 402(1-2): 154-8, 2006 Jul 10.
Article in English | MEDLINE | ID: mdl-16644126

ABSTRACT

It has been reported that inflammatory processes are associated with the pathophysiology of Alzheimer's disease (AD), and the treatment of AD using anti-inflammatory agents slows the progress of AD. Marine algae have been utilized in food products as well as in medicine products for a variety of purposes. In this study, we investigated the neuroprotective effects of methanol extracts of Ulva conglobata (U. conglobata), a marine algae, on glutamate-induced neurotoxicity in the murine hippocampal HT22 cell line and the anti-inflammatory effects on interferon gamma (IFN-gamma)-induced microglial activation in BV2 cells. U. conglobata methanol extracts significantly attenuated the neurotoxicity induced by glutamate in HT22 cells and inhibited nitric oxide production induced by IFN-gamma in BV2 cells. U. conglobata methanol extract treatments were also examined and it was found that they almost completely suppressed the expression of the proinflammatory enzyme cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS). These results suggest that U. conglobata possesses therapeutic potential for combating neurodegenerative diseases associated with neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Hippocampus/cytology , Microglia/drug effects , Neurons/drug effects , Ulva/chemistry , Animals , Blotting, Western/methods , Cell Line , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Glutamic Acid/pharmacology , Interferon-gamma/pharmacology , Mice , Nitric Oxide Synthase Type II/metabolism
17.
J Biol Chem ; 281(1): 617-25, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16253999

ABSTRACT

The appropriate regulation of the actin cytoskeleton is essential for cell movement, changes in cell shape, and formation of membrane protrusions like lamellipodia and filopodia. Moreover, several regulatory proteins affecting actin dynamics have been identified in the motile regions of cells. Here, we provide evidence for the involvement of SPIN90 in the regulation of actin cytoskeleton and actin comet tail formation. SPIN90 was distributed throughout the cytoplasm in COS-7 cells, but exposing the cells to platelet-derived growth factor (PDGF) caused a redistribution of SPIN90 to the cell cortex and the formation of lamellipodia (or membrane ruffles), both of which were dramatically inhibited in SPIN90-knockdown cells. In addition, the binding of the C terminus of SPIN90 with both the Arp2/3 complex (actin-related proteins Arp 2 and Arp 3) and G-actin activates the former, leading to actin polymerization in vitro. And when coexpressed with phosphatidylinositol 4-phosphate 5 kinase, SPIN90 was observed within actin comet tails. Taken these findings suggest that SPIN90 participates in reorganization of the actin cytoskeleton and in actin-based cell motility.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2/metabolism , Actin-Related Protein 3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Muscle Proteins/metabolism , Pseudopodia/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , COS Cells , Cell Movement/physiology , Chlorocebus aethiops , Cricetinae , Humans , Kidney/cytology , Molecular Sequence Data , Muscle Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA, Small Interfering , Transfection
18.
J Neurosci Res ; 82(6): 831-8, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16273545

ABSTRACT

Xanthorrhizol, a natural sesquiterpenoid isolated from the rhizome of Curcuma xanthorrhiza Roxb (Zingiberaceae), has antibacterial activities and protective effects against cisplatin-induced hepatotoxicity. In this study, we investigated the activities of xanthorrhizol as an antioxidant or antiinflammatory agent using neuronal and microglial cells. Xanthorrhizol had potent neuroprotective effects on glutamate-induced neurotoxicity and reactive oxygen species (ROS) generation in the murine hippocampal HT22 cell line. Also, xanthorrhizol inhibited H(2)O(2)-induced lipid peroxidation in rat brain homogenates. The properties of xanthorrhizol as an antiinflammatory agent were investigated in microglial activation by lipopolysaccharide. It reduced the expression of cyclooxygenase-2 and the inducible nitric oxide synthase, which consequently resulted in the reduction of nitric oxide. The production of proinflammatory cytokines, such as interleukin-6 and tumor necrosis factor-alpha in activated microglial cells, was reduced by xanthorrhizol. These results suggest that xanthorrhizol could be an effective candidate for the treatment of Alzheimer's disease- and other neurological disease-related ROS and inflammation.


Subject(s)
Antioxidants/pharmacology , Hippocampus/cytology , Microglia/drug effects , Neurons/drug effects , Phenols/pharmacology , Analysis of Variance , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/chemistry , Blotting, Western/methods , Cell Survival/drug effects , Cells, Cultured , Curcumin/pharmacology , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Hydrogen Peroxide/toxicity , Interleukin-6/metabolism , Lipid Peroxidation/drug effects , Mice , Microglia/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Phenols/chemistry , Rats , Tumor Necrosis Factor-alpha/metabolism
19.
Biochem Biophys Res Commun ; 331(4): 1264-9, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15883012

ABSTRACT

Epidemiological studies suggest that the treatments of anti-inflammatory agents and anti-oxidants slow the progress of neurological diseases. Lignans are anti-oxidants and phytoestrogens found in a variety of plants. In this study, we investigated the neuroprotective effect of macelignan on glutamate-induced neurotoxicity and reactive oxygen species (ROS) in murine hippocampal HT22 cell line. Macelignan significantly attenuated the ROS production and neurotoxicity induced by glutamate in HT22 cell. Also, the properties of macelignan as an anti-inflammatory agent were investigated in microglials activation by lipopolysaccharide (LPS). It potently suppressed the expression of cyclooxygenase-2 and inducible nitric oxide synthase, that consequently resulted in the reduction of nitric oxide in LPS-treated microglial cells. It also significantly suppressed the production of pro-inflammatory cytokine tumor necrosis factor-alpha and interleukin-6. These results suggest that macelignan possesses therapeutic potentials against neurodegenerative diseases with oxidative stress and neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Hippocampus/drug effects , Lignans/pharmacology , Microglia/drug effects , Animals , Cells, Cultured , Hippocampus/cytology , Hippocampus/enzymology , Hippocampus/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Microglia/cytology , Microglia/enzymology , Microglia/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Tumor Necrosis Factor-alpha/metabolism
20.
J Biol Chem ; 278(52): 52116-23, 2003 Dec 26.
Article in English | MEDLINE | ID: mdl-14559906

ABSTRACT

SPIN90 is a widely expressed Nck-binding protein that contains one Src homology 3 (SH3) domain, three Pro-rich motifs, and a serine/threonine-rich region, and is known to participate in sarcomere assembly during cardiac myocyte differentiation. We used in vitro binding assays and yeast two-hybrid screening analysis to identify Nck, betaPIX, Wiscott-Aldrich syndrome protein (WASP), and ERK1 as SPIN90-binding proteins. It appears that betaPIX, WASP, and SPIN90 form a complex that interacts with Nck in a manner dependent upon cell adhesion to extracellular matrix. The betaPIX.WASP.SPIN90.Nck interaction was abolished in suspended and cytochalasin D-treated cells, but was recovered when cells were replated on fibronectin-coated dishes. The SPIN90.betaPIX.WASP complex was stable, even in suspended cells, suggesting SPIN90 serves as an adaptor molecule to recruit other proteins to Nck at focal adhesions. In addition, we found that overexpression of the SPIN90 SH3 domain or Pro-rich region, respectively, abolished SPIN90.Nck and SPIN90.betaPIX interactions, resulting in detachment of cells from extracellular matrix. SPIN90 was phosphorylated by ERK1, which was, itself, activated by cell adhesion and platelet-derived growth factor. Such phosphorylation of SPIN90 likely promotes the interaction of the SPIN90.betaPIX.WASP complex and Nck. It thus appears that the interaction of the betaPIX.WASP.SPIN90 complex with Nck is crucial for stable cell adhesion and can be dynamically modulated by SPIN90 phosphorylation that is dependent on cell adhesion and ERK activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins/metabolism , Oncogene Proteins/metabolism , Amino Acid Motifs , Cell Adhesion , Cell Cycle Proteins/metabolism , Cell Differentiation , Cytochalasin D/pharmacology , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Focal Adhesions , Glutathione Transferase/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Humans , Immunoblotting , Mitogen-Activated Protein Kinase 3 , Models, Genetic , Muscle Cells/metabolism , Phosphorylation , Precipitin Tests , Proline/chemistry , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , Transfection , Two-Hybrid System Techniques , Wiskott-Aldrich Syndrome Protein , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...