Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mitochondrial DNA B Resour ; 9(3): 314-317, 2024.
Article in English | MEDLINE | ID: mdl-38450411

ABSTRACT

Calanthe sieboldii Decne. ex Regel is a terrestrial orchid with high ornamental and commercial value. In the present study, the chloroplast genome of C. sieboldii was characterized using Illumina technology. The chloroplast genome is 158,345 bp in length with a total AT content of 63.28%. There are 127 genes, comprising 37 tRNA genes, 82 protein-coding genes, and 8 rRNA genes. Phylogenetic relationship analysis was performed using common protein-coding genes extracted from 13 chloroplast genomes of Orchidaceae. It was revealed that C. sieboldi was sister to C. hancockii and closely clustered with C. aristulifera and C. henryi. These findings provide valuable genomic resources that are helpful for further phylogenetic and evolutionary studies of Calanthe.

2.
Asian J Surg ; 47(1): 420-424, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37770357

ABSTRACT

BACKGROUND: When tying knots, some surgeons do not pay particular attention to the direction in which they pull to lay down throws. We examine to what extent does pulling direction influence on knot security. METHODS: A total of 368 residents were instructed to tie knots with from 2 to 7 throws using silk braided suture in 3-0 gauge. The direction in which they pulled to lay down throws was recorded. Only the knots tied either by pulling in alternate directions (Group A) or in constant direction (Group C) from the first throw to the last were involved in statistical analysis. Tensile strength and untying rate of the knots were then measured for comparative analysis. RESULTS: For knots with from 2 to 7 throws, the tensile strength of the ones from Group A was significantly higher than that of the ones from Group C (p < 0.05), respectively. For knots with from 5 to 7 throws, the untying rate of the ones from Group A was significantly lower than that of the ones from Group C (p < 0.05), respectively. For the unraveled knots with from 2 to 7 throws (except for the ones with 5 throws), the tensile strength of the ones from Group A was significantly higher than that of the ones from Group C (p < 0.05), respectively. CONCLUSION: Pulling in constant direction results in inferior knot security. Surgeons must ascertain the influence of pulling direction on knot security, and try to achieve superior security with fewer throws to ensure patient safety.


Subject(s)
Suture Techniques , Sutures , Humans , Tensile Strength , Research Design
3.
Int J Syst Evol Microbiol ; 72(10)2022 Oct.
Article in English | MEDLINE | ID: mdl-36282564

ABSTRACT

Three Gram-stain-negative, motile, with amphilophotrichous flagella, and rod-shaped bacteria (LJ1, LJ2T and LJ3) were isolated from lower leaves with black spots on flue-cured tobacco in Yunnan, PR China. The results of phylogenetic analysis based on 16S rRNA gene sequences indicate that all the strains from tobacco were closely related to the type strains of the Pseudomonas syringae group within the P. fluorescens lineage and LJ2T has the highest sequence identities with P. cichorii DSM 50259T (99.92 %), P. capsici Pc19-1T (99.67 %) and P. ovata F51T (98.94 %) . The 16S rRNA gene sequence identities between LJ2T and other members of the genus Pseudomonas were below 98.50%. The average nucleotide identity by blast (ANIb) values between LJ2T and P. cichorii DSM 50259T, P. capsici Pc19-1T and P. ovata F51T were less than 95 %, and the in silico DNA-DNA hybridization (isDDH) values (yielded by formula 2) were less than 70 %. The major fatty acids were C16  :  1ω7c and/or C16  :  1ω6c (summed feature 3), C16  :  0 and C18  :  1ω7c and/or C18  :  1ω6c (summed feature 8). The polar lipids profile of LJ2T consisted of diphosphatidylglycerol, phosphatidylethanolamine, phosphatidylglycerol, phosphatidylcholine, two unidentified phospholipids and one unidentified glycolipid. The predominant respiratory quinone was Q-9. The DNA G+C content of LJ2T was 58.4 mol%. On the basis of these data, we concluded that LJ2T represents a novel species of the genus Pseudomonas, for which the name Pseudomonas lijiangensis sp. nov. is proposed. The type strain of Pseudomonas lijiangensis sp. nov. is LJ2T (=CCTCC AB 2021465T=GDMCC 1.2884T=JCM 35177T).


Subject(s)
Phosphatidylethanolamines , Pseudomonas , RNA, Ribosomal, 16S/genetics , Phylogeny , Base Composition , Nicotiana , DNA, Bacterial/genetics , Cardiolipins , Bacterial Typing Techniques , Fatty Acids/chemistry , Genes, Bacterial , Sequence Analysis, DNA , China , Phospholipids , Phosphatidylcholines , Glycolipids , Quinones , Nucleotides
4.
ACS Chem Neurosci ; 13(10): 1549-1557, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35522720

ABSTRACT

Synaptic and cognitive deficits mediated by a severe reduction in excitatory neurotransmission caused by a disproportionate accumulation of the neuronal protein tau in dendritic spines is a fundamental mechanism that has been found repeatedly in models of tauopathies, including Alzheimer's disease, Lewy body dementia, frontotemporal dementia, and traumatic brain injury. Synapses thus damaged may contribute to dementia, among the most feared cause of debilitation in the elderly, and currently there are no treatments to repair them. Caspase-2 (Casp2) is an essential component of this pathological cascade. Although it is believed that Casp2 exerts its effects by hydrolyzing tau at aspartate-314, forming Δtau314, it is also possible that a noncatalytic mechanism is involved because catalytically dead Casp2 is biologically active in at least one relevant cellular pathway, that is, autophagy. To decipher whether the pathological effects of Casp2 on synaptic function are due to its catalytic or noncatalytic properties, we discovered and characterized a new Casp2 inhibitor, compound 1 [pKi (Casp2) = 8.12], which is 123-fold selective versus Casp3 and >2000-fold selective versus Casp1, Casp6, Casp7, and Casp9. In an in vitro assay based on Casp2-mediated cleavage of tau, compound 1 blocked the production of Δtau314. Importantly, compound 1 prevented tau from accumulating excessively in dendritic spines and rescued excitatory neurotransmission in cultured primary rat hippocampal neurons expressing the P301S tau variant linked to FTDP-17, a familial tauopathy. These results support the further development of small-molecule Casp2 inhibitors to treat synaptic deficits in tauopathies.


Subject(s)
Frontotemporal Dementia , Tauopathies , Animals , Caspase 2/metabolism , Disease Models, Animal , Frontotemporal Dementia/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , Rats , Synaptic Transmission , Tauopathies/metabolism , tau Proteins/metabolism
5.
Biophys J ; 120(16): 3272-3282, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34293301

ABSTRACT

Chronic traumatic encephalopathy is a neurodegenerative disease associated with repeated traumatic brain injury (TBI). Chronic traumatic encephalopathy is a tauopathy, in which cognitive decline is accompanied by the accumulation of neurofibrillary tangles of the protein tau in patients' brains. We recently found that mechanical force alone can induce tau mislocalization to dendritic spines and loss of synaptic function in in vitro neuronal cultures with random cell organization. However, in the brain, neurons are highly aligned, so here we aimed to determine how neuronal organization influences early-stage tauopathy caused by mechanical injury. Using microfabricated cell culture constructs to control the growth of neurites and an in vitro simulated TBI device to apply controlled mechanical deformation, we found that neuronal orientation with respect to the direction of a uniaxial high-strain-rate stretch injury influences the degree of tau pathology in injured neurons. We found that a mechanical stretch applied parallel to the neurite alignment induces greater mislocalization of tau proteins to dendritic spines than does a stretch with the same strain applied perpendicular to the neurites. Synaptic function, characterized by the amplitude of miniature excitatory postsynaptic currents, was similarly decreased in neurons with neurites aligned parallel to stretch, whereas in neurons aligned perpendicular to stretch, it had little to no functional loss. Experimental injury parameters (strain, strain rate, direction of stretch) were combined with a standard viscoelastic solid model to show that in our in vitro model, neurite work density during stretch correlates with tau mislocalization. These findings suggest that in a TBI, the magnitude of brain deformation is not wholly predictive of neurodegenerative consequences of TBI but that deformation relative to local neuronal architecture and the neurite mechanical energy during injury are better metrics for predicting trauma-induced tauopathy.


Subject(s)
Chronic Traumatic Encephalopathy , Neurodegenerative Diseases , Humans , Neurites , Neurofibrillary Tangles , tau Proteins
6.
J Physiol ; 599(9): 2483-2498, 2021 05.
Article in English | MEDLINE | ID: mdl-31194886

ABSTRACT

KEY POINTS: Tau mislocalization to dendritic spines and associated postsynaptic deficits are mediated through different and non-overlapping phosphorylation sites. Tau mislocalization to dendritic spines depends upon the phosphorylation of either Ser396 or Ser404 in the C-terminus. Postsynaptic dysfunction instead depends upon the phosphorylation of at least one of five residues in the proline-rich region of tau. The blockade of both glycogen synthetase kinase 3ß and cyclin-dependent kinase 5 is required to prevent P301L-induced tau mislocalization to dendritic spines, supporting redundant pathways that control tau mislocalization to spines. ABSTRACT: Tau protein consists of an N-terminal projection domain, a microtubule-binding domain and a C-terminal domain. In neurodegenerative diseases, including Alzheimer's disease and frontotemporal dementia, the hyperphosphorylation of tau changes its shape, binding partners and resulting function. An early consequence of tau phosphorylation by proline-directed kinases is postsynaptic dysfunction associated with the mislocalization of tau to dendritic spines. The specific phosphorylation sites leading to these abnormalities have not been elucidated. Here, using imaging and electrophysiological techniques to study cultured rat hippocampal neurons, we show that postsynaptic dysfunction results from a sequential process involving differential phosphorylation in the N-terminal and C-terminal domains. First, tau mislocalizes to dendritic spines, in a manner that depends upon the phosphorylation of either Ser396 or Ser404 in the C-terminal domain. The blockade of both glycogen synthetase kinase 3ß and cyclin-dependent kinase 5 prevents tau mislocalization to dendritic spines. Second, a reduction of functional AMPA receptors depends upon the phosphorylation of at least one of five residues (Ser202, Thr205, Thr212, Thr217 and Thr231) in the proline-rich region of the N-terminal domain. This is the first report of differential phosphorylation in distinct tau domains governing separate, but linked, steps leading to synaptic dysfunction.


Subject(s)
Alzheimer Disease , tau Proteins , Animals , Cells, Cultured , Neurons/metabolism , Phosphorylation , Rats
7.
Proc Natl Acad Sci U S A ; 117(46): 29069-29079, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33139536

ABSTRACT

Chronic traumatic encephalopathy (CTE) is associated with repeated traumatic brain injuries (TBI) and is characterized by cognitive decline and the presence of neurofibrillary tangles (NFTs) of the protein tau in patients' brains. Here we provide direct evidence that cell-scale mechanical deformation can elicit tau abnormalities and synaptic deficits in neurons. Using computational modeling, we find that the early pathological loci of NFTs in CTE brains are regions of high deformation during injury. The mechanical energy associated with high-strain rate deformation alone can induce tau mislocalization to dendritic spines and synaptic deficits in cultured rat hippocampal neurons. These cellular changes are mediated by tau hyperphosphorylation and can be reversed through inhibition of GSK3ß and CDK5 or genetic deletion of tau. Together, these findings identify a mechanistic pathway that directly relates mechanical deformation of neurons to tau-mediated synaptic impairments and provide a possibly exploitable therapeutic pathway to combat CTE.


Subject(s)
Brain Injuries, Traumatic/metabolism , Chronic Traumatic Encephalopathy/metabolism , Dendritic Spines/metabolism , Neurons/metabolism , tau Proteins/metabolism , Animals , Brain/metabolism , Brain Injuries, Traumatic/pathology , Chronic Traumatic Encephalopathy/pathology , Cyclin-Dependent Kinase 5/metabolism , Female , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Humans , Male , Neurofibrillary Tangles/metabolism , Rats , tau Proteins/genetics
8.
Neuroscientist ; 26(5-6): 503-520, 2020.
Article in English | MEDLINE | ID: mdl-32389059

ABSTRACT

Increasingly, research suggests that neurodegenerative diseases and dementias are caused not by unique, solitary cellular mechanisms, but by multiple contributory mechanisms manifesting as heterogeneous clinical presentations. However, diverse neurodegenerative diseases also share common pathological hallmarks and cellular mechanisms. One such mechanism involves the redistribution of the microtubule associated protein tau from the axon into the somatodendritic compartment of neurons, followed by the mislocalization of tau into dendritic spines, resulting in postsynaptic functional deficits. Here we review various signaling pathways that trigger the redistribution of tau to the cell body and dendritic tree, and its mislocalization to dendritic spines. The convergence of multiple pathways in different disease models onto this final common pathway suggests that it may be an attractive pathway to target for developing new treatments for neurodegenerative diseases.


Subject(s)
Dendritic Spines/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Synapses/metabolism , Amyloid beta-Peptides/metabolism , Animals , Humans , tau Proteins/metabolism
9.
Acta Neuropathol ; 138(4): 551-574, 2019 10.
Article in English | MEDLINE | ID: mdl-31168644

ABSTRACT

Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are clinically and neuropathologically highly related α-synucleinopathies that collectively constitute the second leading cause of neurodegenerative dementias. Genetic and neuropathological studies directly implicate α-synuclein (αS) abnormalities in PDD and DLB pathogenesis. However, it is currently unknown how αS abnormalities contribute to memory loss, particularly since forebrain neuronal loss in PDD and DLB is less severe than in Alzheimer's disease. Previously, we found that familial Parkinson's disease-linked human mutant A53T αS causes aberrant localization of the microtubule-associated protein tau to postsynaptic spines in neurons, leading to postsynaptic deficits. Thus, we directly tested if the synaptic and memory deficits in a mouse model of α-synucleinopathy (TgA53T) are mediated by tau. TgA53T mice exhibit progressive memory deficits associated with postsynaptic deficits in the absence of obvious neuropathological and neurodegenerative changes in the hippocampus. Significantly, removal of endogenous mouse tau expression in TgA53T mice (TgA53T/mTau-/-), achieved by mating TgA53T mice to mouse tau-knockout mice, completely ameliorates cognitive dysfunction and concurrent synaptic deficits without affecting αS expression or accumulation of selected toxic αS oligomers. Among the known tau-dependent effects, memory deficits in TgA53T mice were associated with hippocampal circuit remodeling linked to chronic network hyperexcitability. This remodeling was absent in TgA53T/mTau-/- mice, indicating that postsynaptic deficits, aberrant network hyperactivity, and memory deficits are mechanistically linked. Our results directly implicate tau as a mediator of specific human mutant A53T αS-mediated abnormalities related to deficits in hippocampal neurotransmission and suggest a mechanism for memory impairment that occurs as a consequence of synaptic dysfunction rather than synaptic or neuronal loss. We hypothesize that these initial synaptic deficits contribute to network hyperexcitability which, in turn, exacerbate cognitive dysfunction. Our results indicate that these synaptic changes present potential therapeutic targets for amelioration of memory deficits in α-synucleinopathies.


Subject(s)
Memory Disorders/metabolism , Synapses/metabolism , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Transgenic , Neuronal Plasticity , Neurons/metabolism , Neurons/pathology , Synapses/pathology , Synucleinopathies/genetics , Synucleinopathies/pathology , alpha-Synuclein/genetics , tau Proteins/genetics
10.
J Neurosci ; 38(45): 9754-9767, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30249789

ABSTRACT

Abnormalities in α-synuclein are implicated in the pathogenesis of Parkinson's disease (PD). Because α-synuclein is highly concentrated within presynaptic terminals, presynaptic dysfunction has been proposed as a potential pathogenic mechanism. Here, we report novel, tau-dependent, postsynaptic deficits caused by A53T mutant α-synuclein, which is linked to familial PD. We analyzed synaptic activity in hippocampal slices and cultured hippocampal neurons from transgenic mice of either sex expressing human WT, A53T, and A30P α-synuclein. Increased α-synuclein expression leads to decreased spontaneous synaptic vesicle release regardless of genotype. However, only those neurons expressing A53T α-synuclein exhibit postsynaptic dysfunction, including decreased miniature postsynaptic current amplitude and decreased AMPA to NMDA receptor current ratio. We also found that long-term potentiation and spatial learning were impaired by A53T α-synuclein expression. Mechanistically, postsynaptic dysfunction requires glycogen synthase kinase 3ß-mediated tau phosphorylation, tau mislocalization to dendritic spines, and calcineurin-dependent AMPA receptor internalization. Previous studies reveal that human A53T α-synuclein has a high aggregation potential, which may explain the mutation's unique capacity to induce postsynaptic deficits. However, patients with sporadic PD with severe tau pathology are also more likely to have early onset cognitive decline. Our results here show a novel, functional role for tau: mediating the effects of α-synuclein on postsynaptic signaling. Therefore, the unraveled tau-mediated signaling cascade may contribute to the pathogenesis of dementia in A53T α-synuclein-linked familial PD cases, as well as some subgroups of PD cases with extensive tau pathology.SIGNIFICANCE STATEMENT Here, we report mutation-specific postsynaptic deficits that are caused by A53T mutant α-synuclein, which is linked to familial Parkinson's disease (PD). The overexpression of WT, A53T, or A30P human α-synuclein leads to decreased spontaneous synaptic vesicle release. However, only those neurons expressing A53T α-synuclein exhibit tau phosphorylation-dependent postsynaptic dysfunction, which is characterized by decreased miniature postsynaptic current amplitude and decreased AMPA to NMDA receptor current ratio. The mutation-specific postsynaptic effects caused by human A53T α-synuclein will help us better understand the neurobiological basis of this specific form of familial PD. The differential effects of exogenous human WT, A53T, A30P, and E46K α-synuclein on glutamatergic synaptic responses will help to explain the clinical heterogeneity of sporadic and familial PD.


Subject(s)
Mutation/genetics , Neurodegenerative Diseases/genetics , Synaptic Potentials/physiology , alpha-Synuclein/genetics , tau Proteins/genetics , Animals , Animals, Newborn , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiopathology , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/physiopathology , Organ Culture Techniques , Rats , alpha-Synuclein/biosynthesis , tau Proteins/biosynthesis
11.
Eur J Neurosci ; 39(7): 1202-13, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24712999

ABSTRACT

The two histopathological hallmarks of Alzheimer's disease (AD) are amyloid plaques containing multiple forms of amyloid beta (Aß) and neurofibrillary tangles containing phosphorylated tau proteins. As mild cognitive impairment frequently occurs long before the clinical diagnosis of AD, the scientific community has been increasingly interested in the roles of Aß and tau in earlier cellular changes that lead to functional deficits. Therefore, great progress has recently been made in understanding how Aß or tau causes synaptic dysfunction. However, the interaction between the Aß and tau-initiated intracellular cascades that lead to synaptic dysfunction remains elusive. The cornerstone of the two-decade-old hypothetical amyloid cascade model is that amyloid pathologies precede tau pathologies. Although the premise of Aß-tau pathway remains valid, the model keeps evolving as new signaling events are discovered that lead to functional deficits and neurodegeneration. Recent progress has been made in understanding Aß-PrP(C) -Fyn-mediated neurotoxicity and synaptic deficits. Although still elusive, many novel upstream and downstream signaling molecules have been found to modulate tau mislocalization and tau hyperphosphorylation. Here we will discuss the mechanistic interactions between Aß-PrP(C) -mediated neurotoxicity and tau-mediated synaptic deficits in an updated amyloid cascade model with calcium and tau as the central mediators.


Subject(s)
Alzheimer Disease/metabolism , Synapses/metabolism , Synaptic Transmission , tau Proteins/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Humans , Signal Transduction , Synapses/physiology , tau Proteins/genetics
12.
Eur J Neurosci ; 39(7): 1214-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24713000

ABSTRACT

In our previous studies, phosphorylation-dependent tau mislocalization to dendritic spines resulted in early cognitive and synaptic deficits. It is well known that amyloid beta (Aß) oligomers cause synaptic dysfunction by inducing calcineurin-dependent AMPA receptor (AMPAR) internalization. However, it is unknown whether Aß-induced synaptic deficits depend upon tau phosphorylation. It is also unknown whether changes in tau can cause calcineurin-dependent loss of AMPARs in synapses. Here, we show that tau mislocalizes to dendritic spines in cultured hippocampal neurons from APPSwe Alzheimer's disease (AD)-transgenic mice and in cultured rat hippocampal neurons treated with soluble Aß oligomers. Interestingly, Aß treatment also impairs synaptic function by decreasing the amplitude of miniature excitatory postsynaptic currents (mEPSCs). The above tau mislocalization and Aß-induced synaptic impairment are both diminished by the expression of AP tau, indicating that these events require tau phosphorylation. The phosphatase activity of calcineurin is important for AMPAR internalization via dephosphorylation of GluA1 residue S845. The effects of Aß oligomers on mEPSCs are blocked by the calcineurin inhibitor FK506. Aß-induced loss of AMPARs is diminished in neurons from knock-in mice expressing S845A mutant GluA1 AMPA glutamate receptor subunits. This finding suggests that changes in phosphorylation state at S845 are involved in this pathogenic cascade. Furthermore, FK506 rescues deficits in surface AMPAR clustering on dendritic spines in neurons cultured from transgenic mice expressing P301L tau proteins. Together, our results support the role of tau and calcineurin as two intermediate signaling molecules between Aß initiation and eventual synaptic dysfunction early in AD pathogenesis.


Subject(s)
Amyloid beta-Peptides/toxicity , Dendritic Spines/metabolism , Miniature Postsynaptic Potentials , Peptide Fragments/toxicity , Receptors, AMPA/metabolism , tau Proteins/metabolism , Animals , Calcineurin/metabolism , Calcineurin Inhibitors/pharmacology , Cells, Cultured , Dendritic Spines/drug effects , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials , Mice , Mutation , Phosphorylation , Protein Transport , Receptors, AMPA/genetics , Tacrolimus/pharmacology , tau Proteins/genetics
13.
Mol Pharmacol ; 82(2): 333-43, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22596350

ABSTRACT

Drug-induced plasticity of excitatory synapses has been proposed to be the cellular mechanism underlying the aberrant learning associated with addiction. Exposure to various drugs of abuse causes both morphological plasticity of dendritic spines and functional plasticity of excitatory synaptic transmission. Chronic activation of µ-opioid receptors (MOR) in cultured hippocampal neurons causes two forms of synaptic plasticity: loss of dendritic spines and loss of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. With use of live imaging, patch-clamp electrophysiology, and immunocytochemistry, the present study reveals that these two forms of synaptic plasticity are mediated by separate, but interactive, intracellular signaling cascades. The inhibition of Ca(2+)/calmodulin-dependent protein kinase II with 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine (KN-62) blocks MOR-mediated structural plasticity of dendritic spines, but not MOR-mediated cellular redistribution of GluR1 and GluR2 AMPA receptor subunits. In contrast, the inhibition of calcineurin with tacrolimus (FK506) blocks both cellular processes. These findings support the idea that drug-induced structural and functional plasticity of dendritic spines is mediated by divergent, but interactive, signaling pathways.


Subject(s)
Dendritic Spines/chemistry , Dendritic Spines/drug effects , Morphine/pharmacology , Neuronal Plasticity/drug effects , Animals , Animals, Newborn , Cells, Cultured , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/physiology , Neuronal Plasticity/physiology , Neurons/chemistry , Neurons/drug effects , Neurons/physiology , Rats , Structure-Activity Relationship
14.
Neuron ; 68(6): 1067-81, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21172610

ABSTRACT

The microtubule-associated protein tau accumulates in Alzheimer's and other fatal dementias, which manifest when forebrain neurons die. Recent advances in understanding these disorders indicate that brain dysfunction precedes neurodegeneration, but the role of tau is unclear. Here, we show that early tau-related deficits develop not from the loss of synapses or neurons, but rather as a result of synaptic abnormalities caused by the accumulation of hyperphosphorylated tau within intact dendritic spines, where it disrupts synaptic function by impairing glutamate receptor trafficking or synaptic anchoring. Mutagenesis of 14 disease-associated serine and threonine amino acid residues to create pseudohyperphosphorylated tau caused tau mislocalization while creation of phosphorylation-deficient tau blocked the mistargeting of tau to dendritic spines. Thus, tau phosphorylation plays a critical role in mediating tau mislocalization and subsequent synaptic impairment. These data establish that the locus of early synaptic malfunction caused by tau resides in dendritic spines.


Subject(s)
Dendritic Spines/metabolism , Nerve Degeneration/metabolism , Synapses/metabolism , tau Proteins/metabolism , Animals , Cells, Cultured , Dendritic Spines/chemistry , Dendritic Spines/physiology , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Nerve Degeneration/physiopathology , Phosphorylation/physiology , Rats , Synapses/chemistry , Synapses/physiology , tau Proteins/physiology
15.
J Neurosci ; 30(45): 15304-16, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21068335

ABSTRACT

Chronic morphine treatment resulting in the alteration of postsynaptic levels of AMPA receptors, thereby modulating synaptic strength, has been reported. However, the mechanism underlying such drug-induced synaptic modification has not been resolved. By monitoring the GluR1 trafficking in primary hippocampal neurons using the pHluorin-GluR1 imaging and biotinylation studies, we observed that prolonged morphine exposure significantly induced loss of synaptic and extrasynaptic GluR1 by internalization. The morphine-induced GluR1 endocytosis was independent of neural network activities or NMDA receptor activities, as neither blocking the sodium channels with tetrodotoxin nor NMDA receptors with dl-APV altered the effects of morphine. Instead, morphine-induced GluR1 endocytosis is attributed to a change in the phosphorylation state of the GluR1 at Ser(845) as morphine significantly decreased the dephosphorylation of GluR1 at this site. Such changes in Ser(845) phosphorylation required morphine-induced activation of calcineurin, based on the observations that a calcineurin inhibitor, FK506, completely abrogated the dephosphorylation, and morphine treatment led to an increase in calcineurin enzymatic activity, even in the presence of dl-APV. Importantly, pretreatment with FK506 and overexpression of the GluR1 mutants, S845D (phospho-mimic) or S845A (phospho-blocking) attenuated the morphine-induced GluR1 endocytosis. Therefore, the calcineurin-mediated GluR1-S845 dephosphorylation is critical for the morphine-induced changes in the postsynaptic AMPA receptor level. Together, these findings reveal a novel molecular mechanism for opioid-induced neuronal adaptation and/or synaptic impairment.


Subject(s)
Calcineurin/metabolism , Hippocampus/drug effects , Morphine/pharmacology , Neurons/drug effects , Phosphorylation/drug effects , Receptors, AMPA/metabolism , Animals , Blotting, Western , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Microscopy, Confocal , Narcotics/pharmacology , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Synapses/drug effects , Synapses/metabolism
16.
Neuropsychopharmacology ; 34(9): 2097-111, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19295508

ABSTRACT

Fentanyl is a frequently used and abused opioid analgesic and can cause internalization of mu opioid receptors (MORs). Receptor internalization modulates the signaling pathways of opioid receptors. As changes in dendritic spines and synaptic AMPA receptors play important roles in addiction and memory loss, we investigated how fentanyl affects dendritic spines and synaptic AMPA receptors in cultured hippocampal neurons. Fentanyl at low concentrations (0.01 and 0.1 microM) caused the collapse of dendritic spines and decreased the number of AMPA receptor clusters. In contrast, fentanyl at high concentrations (1 and 10 microM) had opposite effects, inducing the emergence of new spines and increasing the number of AMPA receptor clusters. These dose-dependent bidirectional effects of fentanyl were blocked by a selective MOR antagonist CTOP at 5 microM. In neurons that had been transfected with HA-tagged or GFP-tagged MORs, fentanyl at high concentrations induced persistent and robust internalization of MORs, whereas fentanyl at lower concentrations induced little or transient receptor internalization. The blockade of receptor internalization with the expression of dominant-negative Dynamin I (the K44E mutant) reversed the effect of fentanyl at high concentrations, supporting a role of receptor internalization in modulating the dose-dependent effects of fentanyl. In contrast to morphine, the effects of fentanyl on dendritic spines are distinctively bidirectional and concentration dependent, probably due to its ability to induce robust internalization of MORs at high concentrations. The characterization of the effects of fentanyl on spines and AMPA receptors may help us understand the roles of MOR internalization in addiction and cognitive deficits.


Subject(s)
Analgesics, Opioid/pharmacology , Dendritic Spines/drug effects , Fentanyl/pharmacology , Receptors, AMPA/metabolism , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/administration & dosage , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Cells, Cultured , Central Nervous System Agents , Dendritic Spines/physiology , Dose-Response Relationship, Drug , Dynamin I/genetics , Dynamin I/metabolism , Fentanyl/administration & dosage , Hippocampus/drug effects , Hippocampus/physiology , Mutation, Missense , Neurons/drug effects , Neurons/physiology , Protein Transport/drug effects , Rats , Receptors, Opioid, mu/antagonists & inhibitors , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Synapses/drug effects , Synapses/physiology
17.
Mol Cell Neurosci ; 35(3): 456-69, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17513124

ABSTRACT

This study has examined the relationship between the effects of opioids on the internalization of mu opioid receptors (MORs) and the morphology of dendritic spines. Several opioids (morphine, etorphine, DAMGO or methadone) were applied to cultured hippocampal neurons. Live imaging and biochemical techniques were used to examine the dynamic changes in MOR internalization and spine morphology. This study reveals that MOR internalization can regulate opioid-induced morphological changes in dendritic spines: (1) Chronic treatment with morphine, which induced minimal receptor internalization, caused collapse of dendritic spines. In contrast, "internalizing" opioids such as DAMGO and etorphine induced the emergence of new spines. It reveals that opioid-induced changes in spines vary greatly depending on how the applied opioid agonist affects MOR internalization. (2) The blockade of receptor internalization by dominant negative mutant of dynamin, K44E, reversed the effects of DAMGO and etorphine. It indicates that receptor internalization is necessary for the distinct effects of DAMGO and etorphine on spines. (3) In neurons that were cultured from MOR knock-out mice and had been co-transfected with DsRed and MOR-GFP, morphine caused collapse of spines whereas DAMGO induced emergence of new spines, indicating that opioids can alter the structure of spines via postsynaptic MORs. (4) Methadone at a low concentration induced minimal internalization and had effects that were similar to morphine. At a high concentration, methadone induced robust internalization and had effects that are opposite to morphine. The concentration-dependent opioid-induced changes in dendritic spines might also contribute to the variation in the effects of individual opioids.


Subject(s)
Analgesics, Opioid/pharmacology , Dendritic Spines/drug effects , Neurons/cytology , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/classification , Animals , Animals, Newborn , Cells, Cultured , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Mice , Nonlinear Dynamics , Protein Transport/drug effects , Rats , Time Factors
18.
J Neurophysiol ; 97(2): 1485-94, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17122315

ABSTRACT

Although chronic treatment with morphine is known to alter the function and morphology of excitatory synapses, the effects of other opioids on these synapses are not clear. Here we report distinct effects of several opioids (morphine, [d-ala(2),me-phe(4),gly(5)-ol]enkephalin (DAMGO), and etorphine) on miniature excitatory postsynaptic currents (mEPSCs) in cultured hippocampal neurons: 1) chronic treatment with morphine for >3 days decreased the amplitude, frequency, rise time and decay time of mEPSCs. In contrast, "internalizing" opioids such as etorphine and DAMGO increased the frequency of mEPSCs and had no significant effect on the amplitude and kinetics of mEPSCs. These results demonstrate that different opioids can have distinct effects on the function of excitatory synapses. 2) mu opioid receptor fused with green fluorescence protein (MOR-GFP) is clustered in dendritic spines in most hippocampal neurons but is concentrated in axon-like processes in striatal and corticostriatal nonspiny neurons. It suggests that MORs might mediate pre- or postsynaptic effects depending on cell types. 3) Neurons were cultured from MOR knock-out mice and were exogenously transfected with MOR-GFP. Chronic treatment with morphine suppressed mEPSCs only in neurons that contained postsynaptic MOR-GFP, indicating that opioids can modulate excitatory synaptic transmission postsynaptically. 4) Morphine acutely decreased mEPSC amplitude in neurons expressing exogenous MOR-GFP but had no effect on neurons expressing GFP. It indicates that the low level of endogenous MORs could only allow slow opioid-induced plasticity of excitatory synapses under normal conditions. 5) A theoretical model suggests that morphine might affect the function of spines by decreasing the electrotonic distance from synaptic inputs to the soma.


Subject(s)
Analgesics, Opioid/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , Neurons/drug effects , Animals , Cells, Cultured , Dendrites/drug effects , Electrophysiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Etorphine/pharmacology , Green Fluorescent Proteins/pharmacology , Hippocampus/cytology , Image Processing, Computer-Assisted , Mice , Morphine/pharmacology , Neostriatum/cytology , Neostriatum/drug effects , Neostriatum/physiology , Rats , Stimulation, Chemical , Transfection
19.
J Neurosci ; 26(9): 2380-90, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510715

ABSTRACT

The targeting and surface expression of membrane proteins are critical to their functions. In neurons, synaptic targeting and surface expression of AMPA-type glutamate receptors were found to be critical for synaptic plasticity such as long-term potentiation and long-term depression (LTD). PICK1 (protein interacting with C kinase 1) is a cytosolic protein that interacts with many membrane proteins, including AMPA receptors via its PDZ (postsynaptic density-95/Discs large/zona occludens-1) domain. Its interactions with membrane proteins regulate their subcellular targeting and surface expression. However, the mechanism by which PICK1 regulates protein trafficking has not been fully elucidated. Here, we show that PICK1 directly binds to lipids, mainly phosphoinositides, via its BAR (Bin/amphiphysin/Rvs) domain. Lipid binding of the PICK1 BAR domain is positively regulated by its PDZ domain and negatively regulated by its C-terminal acidic domain. Mutation of critical residues of the PICK1 BAR domain eliminates its lipid-binding capability. Lipid binding of PICK1 controls the subcellular localization of the protein, because BAR domain mutant of PICK1 has diminished synaptic targeting compared with wild-type PICK1. In addition, the BAR domain mutant of PICK1 does not cluster AMPA receptors. Moreover, wild-type PICK1 enhances synaptic targeting of AMPA receptors, whereas the BAR domain mutant of PICK1 fails to do so. The BAR domain mutant of PICK1 loses its ability to regulate surface expression of the AMPA receptors and impairs expression of LTD in hippocampal neurons. Together, our findings indicate that the lipid binding of the PICK1 BAR domain is important for its synaptic targeting, AMPA receptor trafficking, and synaptic plasticity.


Subject(s)
Carrier Proteins/metabolism , Lipids/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Nuclear Proteins/metabolism , Receptors, AMPA/metabolism , Animals , Animals, Newborn , Biotinylation/methods , Blotting, Western/methods , Carrier Proteins/genetics , Cells, Cultured , Chlorocebus aethiops , Cloning, Molecular/methods , Cytoskeletal Proteins , Disks Large Homolog 4 Protein , Dose-Response Relationship, Drug , Electric Stimulation/methods , Embryo, Mammalian , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , Immunoprecipitation/methods , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Long-Term Synaptic Depression/physiology , Long-Term Synaptic Depression/radiation effects , Membrane Proteins/metabolism , Models, Biological , Mutation , Nuclear Proteins/genetics , Patch-Clamp Techniques/methods , Phosphatidylinositols/pharmacokinetics , Protein Binding , Protein Structure, Tertiary/physiology , Protein Transport/physiology , Rats , Subcellular Fractions/drug effects , Subcellular Fractions/physiology , Transfection/methods , Two-Hybrid System Techniques
20.
J Neurosci ; 25(46): 10627-36, 2005 Nov 16.
Article in English | MEDLINE | ID: mdl-16291935

ABSTRACT

Glutamatergic synapses switch from nonspiny synapses to become dendritic spines during early neuronal development. Here, we report that the lack of sufficient Rac1, a small RhoGTPase, contributes to the absence of spinogenesis in immature neurons. The overexpression of green fluorescence protein-tagged wild-type Rac1 initiated the formation of dendritic spines in cultured dissociated hippocampal neurons younger than 11 d in vitro, indicating that Rac1 is likely one of the missing pieces responsible for the lack of spines in immature neurons. The overexpression of wild-type Rac1 also induced the clustering of AMPA receptors (AMPARs) and increased the amplitude of miniature EPSCs (mEPSCs). The expression of constitutively active Rac1 induced the formation of unusually large synapses with large amounts of AMPAR clusters. Also, our live imaging experiments revealed that the contact of an axon induced the clustering of Rac1, and subsequent morphological changes led to spinogenesis. Additionally, the overexpression of wild-type Rac1 and constitutively active Rac1 increased the size of preexisting spines and the amplitude of mEPSCs in mature neurons (>21 d in vitro) within 24 h after transfection. Together, these results indicate that activation of Rac1 enhances excitatory synaptic transmission by recruiting AMPARs to synapses during spinogenesis, thus providing a mechanistic link between presynaptic and postsynaptic developmental changes. Furthermore, we show that Rac1 has two distinct roles at different stages of neuronal development. The activation of Rac1 initiates spinogenesis at an early stage and regulates the function and morphology of preexisting spines at a later stage.


Subject(s)
Dendritic Spines/metabolism , Receptor Aggregation/physiology , Receptors, AMPA/biosynthesis , rac1 GTP-Binding Protein/physiology , Animals , Animals, Newborn , Cells, Cultured , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Hippocampus/cytology , Hippocampus/metabolism , Rats , Receptor Aggregation/genetics , Receptors, AMPA/genetics , Receptors, AMPA/ultrastructure , Synaptic Transmission/genetics , Synaptic Transmission/physiology , rac1 GTP-Binding Protein/biosynthesis , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...