Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
Add more filters

Publication year range
1.
Mol Cell ; 74(3): 466-480.e4, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30930055

ABSTRACT

The mTOR pathway integrates both extracellular and intracellular signals and serves as a central regulator of cell metabolism, growth, survival, and stress responses. Neurotropic viruses, such as herpes simplex virus-1 (HSV-1), also rely on cellular AKT-mTORC1 signaling to achieve viral latency. Here, we define a novel genotoxic response whereby spatially separated signals initiated by extracellular neurotrophic factors and nuclear DNA damage are integrated by the AKT-mTORC1 pathway. We demonstrate that endogenous DNA double-strand breaks (DSBs) mediated by Topoisomerase 2ß-DNA cleavage complex (TOP2ßcc) intermediates are required to achieve AKT-mTORC1 signaling and maintain HSV-1 latency in neurons. Suppression of host DNA-repair pathways that remove TOP2ßcc trigger HSV-1 reactivation. Moreover, perturbation of AKT phosphorylation dynamics by downregulating the PHLPP1 phosphatase led to AKT mis-localization and disruption of DSB-induced HSV-1 reactivation. Thus, the cellular genome integrity and environmental inputs are consolidated and co-opted by a latent virus to balance lifelong infection with transmission.


Subject(s)
DNA Topoisomerases, Type II/genetics , Herpesvirus 1, Human/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Virus Latency/genetics , Animals , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair/genetics , DNA Repair/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Herpesvirus 1, Human/pathogenicity , Humans , MRE11 Homologue Protein/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Neurons/metabolism , Neurons/virology , Phosphorylation , Rats , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
2.
EMBO Rep ; 23(2): e53543, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34842321

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a powerful technique for dissecting the complexity of normal and diseased tissues, enabling characterization of cell diversity and heterogeneous phenotypic states in unprecedented detail. However, this technology has been underutilized for exploring the interactions between the host cell and viral pathogens in latently infected cells. Herein, we use scRNA-seq and single-molecule sensitivity fluorescent in situ hybridization (smFISH) technologies to investigate host single-cell transcriptome changes upon the reactivation of a human neurotropic virus, herpes simplex virus-1 (HSV-1). We identify the stress sensor growth arrest and DNA damage-inducible 45 beta (Gadd45b) as a critical antiviral host factor that regulates HSV-1 reactivation events in a subpopulation of latently infected primary neurons. We show that distinct subcellular localization of Gadd45b correlates with the viral late gene expression program, as well as the expression of the viral transcription factor, ICP4. We propose that a hallmark of a "successful" or "aborted" HSV-1 reactivation state in primary neurons is determined by a unique subcellular localization signature of the stress sensor Gadd45b.


Subject(s)
Antigens, Differentiation/metabolism , Herpesvirus 1, Human , Neurons/virology , Virus Activation , Virus Latency , Gene Expression Regulation , Herpesvirus 1, Human/physiology , Humans , In Situ Hybridization, Fluorescence , Transcriptome
3.
J Neurosci ; 42(19): 3919-3930, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35361702

ABSTRACT

The molecular mechanisms underlying somatodendritic dopamine (DA) release remain unresolved, despite the passing of decades since its discovery. Our previous work showed robust release of somatodendritic DA in submillimolar extracellular Ca2+ concentration ([Ca2+]o). Here we tested the hypothesis that the high-affinity Ca2+ sensor synaptotagmin 7 (Syt7), is a key determinant of somatodendritic DA release and its Ca2+ dependence. Somatodendritic DA release from SNc DA neurons was assessed using whole-cell recording in midbrain slices from male and female mice to monitor evoked DA-dependent D2 receptor-mediated inhibitory currents (D2ICs). Single-cell application of an antibody to Syt7 (Syt7 Ab) decreased pulse train-evoked D2ICs, revealing a functional role for Syt7. The assessment of the Ca2+ dependence of pulse train-evoked D2ICs confirmed robust DA release in submillimolar [Ca2+]o in wild-type (WT) neurons, but loss of this sensitivity with intracellular Syt7 Ab or in Syt7 knock-out (KO) mice. In millimolar [Ca2+]o, pulse train-evoked D2ICs in Syt7 KOs showed a greater reduction in decreased [Ca2+]o than seen in WT mice; the effect on single pulse-evoked DA release, however, did not differ between genotypes. Single-cell application of a Syt1 Ab had no effect on train-evoked D2ICs in WT SNc DA neurons, but did cause a decrease in D2IC amplitude in Syt7 KOs, indicating a functional substitution of Syt1 for Syt7. In addition, Syt1 Ab decreased single pulse-evoked D2ICs in WT cells, indicating the involvement of Syt1 in tonic DA release. Thus, Syt7 and Syt1 play complementary roles in somatodendritic DA release from SNc DA neurons.SIGNIFICANCE STATEMENT The respective Ca2+ dependence of somatodendritic and axonal dopamine (DA) release differs, resulting in the persistence of somatodendritic DA release in submillimolar Ca2+ concentrations too low to support axonal release. We demonstrate that synaptotagmin7 (Syt7), a high-affinity Ca2+ sensor, underlies phasic somatodendritic DA release and its Ca2+ sensitivity in the substantia nigra pars compacta. In contrast, we found that synaptotagmin 1 (Syt1), the Ca2+ sensor underlying axonal DA release, plays a role in tonic, but not phasic, somatodendritic DA release in wild-type mice. However, Syt1 can facilitate phasic DA release after Syt7 deletion. Thus, we show that both Syt1 and Syt7 act as Ca2+ sensors subserving different aspects of somatodendritic DA release processes.


Subject(s)
Dopamine , Substantia Nigra , Synaptotagmin I , Synaptotagmins , Animals , Dendrites , Dopamine/pharmacology , Dopaminergic Neurons , Electric Stimulation , Female , Male , Mice , Synaptotagmin I/genetics , Synaptotagmins/genetics
4.
J Neurosci ; 42(23): 4725-4736, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35577554

ABSTRACT

Physical exercise improves motor performance in individuals with Parkinson's disease and elevates mood in those with depression. Although underlying factors have not been identified, clues arise from previous studies showing a link between cognitive benefits of exercise and increases in brain-derived neurotrophic factor (BDNF). Here, we investigated the influence of voluntary wheel-running exercise on BDNF levels in the striatum of young male wild-type (WT) mice, and on the striatal release of a key motor-system transmitter, dopamine (DA). Mice were allowed unlimited access to a freely rotating wheel (runners) or a locked wheel (controls) for 30 d. Electrically evoked DA release was quantified in ex vivo corticostriatal slices from these animals using fast-scan cyclic voltammetry. We found that exercise increased BDNF levels in dorsal striatum (dStr) and increased DA release in dStr and in nucleus accumbens core and shell. Increased DA release was independent of striatal acetylcholine (ACh), and persisted after a week of rest. We tested a role for BDNF in the influence of exercise on DA release using mice that were heterozygous for BDNF deletion (BDNF+/-). In contrast to WT mice, evoked DA release did not differ between BDNF+/- runners and controls. Complementary pharmacological studies using a tropomyosin receptor kinase B (TrkB) agonist in WT mouse slices showed that TrkB receptor activation also increased evoked DA release throughout striatum in an ACh-independent manner. Together, these data support a causal role for BDNF in exercise-enhanced striatal DA release and provide mechanistic insight into the beneficial effects of exercise in neuropsychiatric disorders, including Parkinson's, depression, and anxiety.SIGNIFICANCE STATEMENT Exercise has been shown to improve movement and cognition in humans and rodents. Here, we report that voluntary exercise for 30 d leads to an increase in evoked DA release throughout the striatum and an increase in BDNF in the dorsal (motor) striatum. The increase in DA release appears to require BDNF, indicated by the absence of DA release enhancement with running in BDNF+/- mice. Activation of BDNF receptors using a pharmacological agonist was also shown to boost DA release. Together, these data support a necessary and sufficient role for BDNF in exercise-enhanced DA release and provide mechanistic insight into the reported benefits of exercise in individuals with dopamine-linked neuropsychiatric disorders, including Parkinson's disease and depression.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dopamine , Parkinson Disease , Acetylcholine/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Corpus Striatum , Dopamine/physiology , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens
5.
Nature ; 520(7548): 499-504, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25874674

ABSTRACT

Oxytocin is important for social interactions and maternal behaviour. However, little is known about when, where and how oxytocin modulates neural circuits to improve social cognition. Here we show how oxytocin enables pup retrieval behaviour in female mice by enhancing auditory cortical pup call responses. Retrieval behaviour required the left but not right auditory cortex, was accelerated by oxytocin in the left auditory cortex, and oxytocin receptors were preferentially expressed in the left auditory cortex. Neural responses to pup calls were lateralized, with co-tuned and temporally precise excitatory and inhibitory responses in the left cortex of maternal but not pup-naive adults. Finally, pairing calls with oxytocin enhanced responses by balancing the magnitude and timing of inhibition with excitation. Our results describe fundamental synaptic mechanisms by which oxytocin increases the salience of acoustic social stimuli. Furthermore, oxytocin-induced plasticity provides a biological basis for lateralization of auditory cortical processing.


Subject(s)
Auditory Cortex/physiology , Maternal Behavior/physiology , Neural Inhibition/physiology , Oxytocin/metabolism , Acoustic Stimulation , Animals , Animals, Newborn , Auditory Cortex/cytology , Auditory Perception/physiology , Evoked Potentials, Auditory , Female , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Receptors, Oxytocin/metabolism , Sexual Abstinence , Vocalization, Animal
6.
Mol Cell Neurosci ; 99: 103395, 2019 09.
Article in English | MEDLINE | ID: mdl-31422108

ABSTRACT

BACE1 is a transmembrane aspartic protease that cleaves various substrates and it is required for normal brain function. BACE1 expression is high during early development, but it is reduced in adulthood. Under conditions of stress and injury, BACE1 levels are increased; however, the underlying mechanisms that drive BACE1 elevation are not well understood. One mechanism associated with brain injury is the activation of injurious p75 neurotrophin receptor (p75), which can trigger pathological signals. Here we report that within 72 h after controlled cortical impact (CCI) or laser injury, BACE1 and p75 are increased and tightly co-expressed in cortical neurons of mouse brain. Additionally, BACE1 is not up-regulated in p75 null mice in response to focal cortical injury, while p75 over-expression results in BACE1 augmentation in HEK-293 and SY5Y cell lines. A luciferase assay conducted in SY5Y cell line revealed that BACE1 expression is regulated at the transcriptional level in response to p75 transfection. Interestingly, this effect does not appear to be dependent upon p75 ligands including mature and pro-neurotrophins. In addition, BACE1 activity on amyloid precursor protein (APP) is enhanced in SY5Y-APP cells transfected with a p75 construct. Lastly, we found that the activation of c-jun n-terminal kinase (JNK) by p75 contributes to BACE1 up-regulation. This study explores how two injury-induced molecules are intimately connected and suggests a potential link between p75 signaling and the expression of BACE1 after brain injury.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain Injuries, Traumatic/metabolism , Receptor, Nerve Growth Factor/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/metabolism , HEK293 Cells , Humans , MAP Kinase Kinase 4/metabolism , Male , Mice , Receptor, Nerve Growth Factor/genetics , Signal Transduction , Up-Regulation
7.
Genes Dev ; 26(14): 1527-32, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22802527

ABSTRACT

Latent herpes simplex virus-1 (HSV1) genomes in peripheral nerve ganglia periodically reactivate, initiating a gene expression program required for productive replication. Whether molecular cues detected by axons can be relayed to cell bodies and harnessed to regulate latent genome expression in neuronal nuclei is unknown. Using a neuron culture model, we found that inhibiting mTOR, depleting its regulatory subunit raptor, or inducing hypoxia all trigger reactivation. While persistent mTORC1 activation suppressed reactivation, a mutant 4E-BP (eIF4E-binding protein) translational repressor unresponsive to mTORC1 stimulated reactivation. Finally, inhibiting mTOR in axons induced reactivation. Thus, local changes in axonal mTOR signaling that control translation regulate latent HSV1 genomes in a spatially segregated compartment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Axons/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , TOR Serine-Threonine Kinases/metabolism , Virus Latency/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Axons/virology , Cells, Cultured , Genome, Viral/physiology , Herpes Simplex/genetics , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Proteins/genetics , Proteins/metabolism , Rats , TOR Serine-Threonine Kinases/genetics
8.
Neurobiol Dis ; 132: 104540, 2019 12.
Article in English | MEDLINE | ID: mdl-31349032

ABSTRACT

INTRODUCTION: Downregulation of brain-derived neurotrophic factor (BDNF) and its cognate neurotrophin receptor, TrkB, were observed during the progression of dementia, but whether the Alzheimer's disease (AD) pathological lesions diffuse plaques, (DPs), neuritic plaques (NPs), and neurofibrillary tangles (NFTs) are related to this alteration remains to be clarified. METHODS: Negative binomial (NB) regressions were performed using gene expression data accrued from a single population of CA1 pyramidal neurons and regional hippocampal dissections obtained from participants in the Rush Religious Orders Study (RROS). RESULTS: Downregulation of Bdnf is independently associated with increased entorhinal cortex NPs. Downregulation of TrkB is independently associated with increased entorhinal cortex NFTs and CA1 NPs during the progression of AD. DISCUSSION: Results indicate that BDNF and TrkB dysregulation contribute to AD neuropathology, most notably hippocampal NPs and NFTs. These data suggest attenuating BDNF/TrkB signaling deficits either at the level of BDNF, TrkB, or downstream of TrkB signaling may abrogate NPs and/or NFTs.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Hippocampus/metabolism , Membrane Glycoproteins/biosynthesis , Neurofibrillary Tangles/metabolism , Plaque, Amyloid/metabolism , Receptor, trkB/biosynthesis , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain-Derived Neurotrophic Factor/genetics , Female , Gene Expression , Hippocampus/pathology , Humans , Male , Membrane Glycoproteins/genetics , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/pathology , Predictive Value of Tests , Receptor, trkB/genetics
9.
Hippocampus ; 29(5): 422-439, 2019 05.
Article in English | MEDLINE | ID: mdl-28888073

ABSTRACT

Hippocampal CA1 pyramidal neurons, a major component of the medial temporal lobe memory circuit, are selectively vulnerable during the progression of Alzheimer's disease (AD). The cellular mechanism(s) underlying degeneration of these neurons and the relationship to cognitive performance remains largely undefined. Here, we profiled neurotrophin and neurotrophin receptor gene expression within microdissected CA1 neurons along with regional hippocampal dissections from subjects who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), or AD using laser capture microdissection (LCM), custom-designed microarray analysis, and qPCR of CA1 subregional dissections. Gene expression levels were correlated with cognitive test scores and AD neuropathology criteria. We found a significant downregulation of several neurotrophin genes (e.g., Gdnf, Ngfb, and Ntf4) in CA1 pyramidal neurons in MCI compared to NCI and AD subjects. In addition, the neurotrophin receptor transcripts TrkB and TrkC were decreased in MCI and AD compared to NCI. Regional hippocampal dissections also revealed select neurotrophic gene dysfunction providing evidence for vulnerability within the hippocampus proper during the progression of dementia. Downregulation of several neurotrophins of the NGF family and cognate neurotrophin receptor (TrkA, TrkB, and TrkC) genes correlated with antemortem cognitive measures including the Mini-Mental State Exam (MMSE), a composite global cognitive score (GCS), and Episodic, Semantic, and Working Memory, Perceptual Speed, and Visuospatial domains. Significant correlations were found between select neurotrophic expression downregulation and neuritic plaques (NPs) and neurofibrillary tangles (NFTs), but not diffuse plaques (DPs). These data suggest that dysfunction of neurotrophin signaling complexes have profound negative sequelae within vulnerable hippocampal cell types, which play a role in mnemonic and executive dysfunction during the progression of AD.


Subject(s)
Alzheimer Disease/pathology , Cognitive Dysfunction/pathology , Hippocampus/pathology , Nerve Growth Factors/metabolism , Pyramidal Cells/pathology , Receptors, Nerve Growth Factor/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Cognitive Dysfunction/metabolism , Disease Progression , Female , Hippocampus/metabolism , Humans , Male , Pyramidal Cells/metabolism
10.
Neuroendocrinology ; 109(3): 277-284, 2019.
Article in English | MEDLINE | ID: mdl-30572337

ABSTRACT

Behavioral choices made by the brain during stress depend on glucocorticoid and brain-derived neurotrophic factor (BDNF) signaling pathways acting in synchrony in the mesolimbic (reward) and corticolimbic (emotion) neural networks. Deregulated expression of BDNF and glucocorticoid receptors in brain valuation areas may compromise the integration of signals. Glucocorticoid receptor phosphorylation upon BDNF signaling in neurons represents one mechanism underlying the integration of BDNF and glucocorticoid signals that when off balance may lay the foundation of maladaptations to stress. Here, we propose that BDNF signaling conditions glucocorticoid responses impacting neural plasticity in the mesocorticolimbic system. This provides a novel molecular framework for understanding how brain networks use BDNF and glucocorticoid signaling contingencies to forge receptive neuronal fields in temporal domains defined by behavioral experience, and in mood disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Glucocorticoids/metabolism , Animals , Humans , Neural Pathways/metabolism , Neurons/metabolism , Signal Transduction
11.
J Biol Chem ; 292(40): 16594-16604, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28821608

ABSTRACT

The function of protein products generated from intramembraneous cleavage by the γ-secretase complex is not well defined. The γ-secretase complex is responsible for the cleavage of several transmembrane proteins, most notably the amyloid precursor protein that results in Aß, a transmembrane (TM) peptide. Another protein that undergoes very similar γ-secretase cleavage is the p75 neurotrophin receptor. However, the fate of the cleaved p75 TM domain is unknown. p75 neurotrophin receptor is highly expressed during early neuronal development and regulates survival and process formation of neurons. Here, we report that the p75 TM can stimulate the phosphorylation of TrkB (tyrosine kinase receptor B). In vitro phosphorylation experiments indicated that a peptide representing p75 TM increases TrkB phosphorylation in a dose- and time-dependent manner. Moreover, mutagenesis analyses revealed that a valine residue at position 264 in the rat p75 neurotrophin receptor is necessary for the ability of p75 TM to induce TrkB phosphorylation. Because this residue is just before the γ-secretase cleavage site, we then investigated whether the p75(αγ) peptide, which is a product of both α- and γ-cleavage events, could also induce TrkB phosphorylation. Experiments using TM domains from other receptors, EGFR and FGFR1, failed to stimulate TrkB phosphorylation. Co-immunoprecipitation and biochemical fractionation data suggested that p75 TM stimulates TrkB phosphorylation at the cell membrane. Altogether, our results suggest that TrkB activation by p75(αγ) peptide may be enhanced in situations where the levels of the p75 receptor are increased, such as during brain injury, Alzheimer's disease, and epilepsy.


Subject(s)
Cell Membrane/metabolism , Membrane Glycoproteins/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkB/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Substitution , Animals , Brain Injuries/genetics , Brain Injuries/metabolism , Cell Membrane/genetics , Epilepsy/genetics , Epilepsy/metabolism , Humans , Membrane Glycoproteins/genetics , Mutagenesis , Mutation, Missense , Phosphorylation , Protein Domains , Rats , Receptor, Nerve Growth Factor/genetics , Receptor, trkB/genetics , Sf9 Cells , Spodoptera
12.
J Neurochem ; 144(3): 302-317, 2018 02.
Article in English | MEDLINE | ID: mdl-28869759

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive deposition of amyloid beta (Aß) and dysregulation of neurotrophic signaling, causing synaptic dysfunction, loss of memory, and cell death. The expression of p75 neurotrophin receptor is elevated in the brain of AD patients, suggesting its involvement in this disease. However, the exact mechanism of its action is not yet clear. Here, we show that p75 interacts with beta-site amyloid precursor protein cleaving enzyme-1 (BACE1), and this interaction is enhanced in the presence of Aß. Our results suggest that the colocalization of BACE1 and amyloid precursor protein (APP) is increased in the presence of both Aß and p75 in cortical neurons. In addition, the localization of APP and BACE1 in early endosomes is increased in the presence of Aß and p75. An increased phosphorylation of APP-Thr668 and BACE1-Ser498 by c-Jun N-terminal kinase (JNK) in the presence of Aß and p75 could be responsible for this localization. In conclusion, our study proposes a potential involvement in amyloidogenesis for p75, which may represent a future therapeutic target for AD. Cover Image for this Issue: doi. 10.1111/jnc.14163.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/metabolism , Endosomes/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Mice, Knockout , Primary Cell Culture , Receptors, Nerve Growth Factor/genetics , Signal Transduction
13.
J Cell Sci ; 129(9): 1866-77, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26966186

ABSTRACT

Proper development of the nervous system requires a temporally and spatially orchestrated set of events including differentiation, synapse formation and neurotransmission. Nerve growth factor (NGF) acting through the TrkA neurotrophin receptor (also known as NTRK1) regulates many of these events. However, the molecular mechanisms responsible for NGF-regulated secretion are not completely understood. Here, we describe a new signaling pathway involving TrkA, ARMS (also known as Kidins220), synembryn-B and Rac1 in NGF-mediated secretion in PC12 cells. Whereas overexpression of ARMS blocked NGF-mediated secretion, without affecting basal secretion, a decrease in ARMS resulted in potentiation. Similar effects were observed with synembryn-B, a protein that interacts directly with ARMS. Downstream of ARMS and synembryn-B are Gαq and Trio proteins, which modulate the activity of Rac1 in response to NGF. Expression of dominant-negative Rac1 rescued the secretion defects of cells overexpressing ARMS or synembryn-B. Thus, this neurotrophin pathway represents a new mechanism responsible for NGF-regulated secretion.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Membrane Proteins/metabolism , Nerve Growth Factor/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Animals , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Nerve Growth Factor/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , PC12 Cells , Phosphoproteins/genetics , Rats , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
14.
Proc Natl Acad Sci U S A ; 112(51): 15737-42, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26630005

ABSTRACT

Neurotrophins and glucocorticoids are robust synaptic modifiers, and deregulation of their activities is a risk factor for developing stress-related disorders. Low levels of brain-derived neurotrophic factor (BDNF) increase the desensitization of glucocorticoid receptors (GR) and vulnerability to stress, whereas higher levels of BDNF facilitate GR-mediated signaling and the response to antidepressants. However, the molecular mechanism underlying neurotrophic-priming of GR function is poorly understood. Here we provide evidence that activation of a TrkB-MAPK pathway, when paired with the deactivation of a GR-protein phosphatase 5 pathway, resulted in sustained GR phosphorylation at BDNF-sensitive sites that is essential for the transcription of neuronal plasticity genes. Genetic strategies that disrupted GR phosphorylation or TrkB signaling in vivo impaired the neuroplasticity to chronic stress and the effects of the antidepressant fluoxetine. Our findings reveal that the coordinated actions of BDNF and glucocorticoids promote neuronal plasticity and that disruption in either pathway could set the stage for the development of stress-induced psychiatric diseases.


Subject(s)
Antidepressive Agents/pharmacology , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/physiology , Signal Transduction/physiology , Stress, Psychological/physiopathology , Animals , Brain-Derived Neurotrophic Factor/physiology , Female , Fluoxetine/pharmacology , MAP Kinase Signaling System , Membrane Glycoproteins/physiology , Mice , Neuronal Plasticity/drug effects , Phosphorylation , Protein-Tyrosine Kinases/physiology , Rats , Rats, Sprague-Dawley , Receptor, trkB
15.
J Neurosci ; 36(8): 2517-35, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26911697

ABSTRACT

Oxytocin is a neuropeptide important for social behaviors such as maternal care and parent-infant bonding. It is believed that oxytocin receptor signaling in the brain is critical for these behaviors, but it is unknown precisely when and where oxytocin receptors are expressed or which neural circuits are directly sensitive to oxytocin. To overcome this challenge, we generated specific antibodies to the mouse oxytocin receptor and examined receptor expression throughout the brain. We identified a distributed network of female mouse brain regions for maternal behaviors that are especially enriched for oxytocin receptors, including the piriform cortex, the left auditory cortex, and CA2 of the hippocampus. Electron microscopic analysis of the cerebral cortex revealed that oxytocin receptors were mainly expressed at synapses, as well as on axons and glial processes. Functionally, oxytocin transiently reduced synaptic inhibition in multiple brain regions and enabled long-term synaptic plasticity in the auditory cortex. Thus modulation of inhibition may be a general mechanism by which oxytocin can act throughout the brain to regulate parental behaviors and social cognition.


Subject(s)
Auditory Cortex/metabolism , Cognition/physiology , Nerve Net/metabolism , Receptors, Oxytocin/biosynthesis , Social Behavior , Amino Acid Sequence , Animals , Auditory Cortex/chemistry , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nerve Net/chemistry , Receptors, Oxytocin/analysis , Receptors, Oxytocin/genetics
16.
Neurobiol Dis ; 97(Pt B): 73-79, 2017 01.
Article in English | MEDLINE | ID: mdl-27015693

ABSTRACT

Growth factor withdrawal has been studied across different species and has been shown to have dramatic consequences on cell survival. In the nervous system, withdrawal of nerve growth factor (NGF) from sympathetic and sensory neurons results in substantial neuronal cell death, signifying a requirement for NGF for the survival of neurons in the peripheral nervous system (PNS). In contrast to the PNS, withdrawal of central nervous system (CNS) enriched brain-derived neurotrophic factor (BDNF) has little effect on cell survival but is indispensible for synaptic plasticity. Given that most early events in neuropsychiatric disorders are marked by a loss of synapses, lack of BDNF may thus be an important part of a cascade of events that leads to neuronal degeneration. Here we review reports on the effects of BDNF withdrawal on CNS neurons and discuss the relevance of the loss in disease.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Central Nervous System Diseases/metabolism , Central Nervous System/metabolism , Neurons/metabolism , Animals , Humans
17.
Clin Sci (Lond) ; 131(1): 13-23, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27908981

ABSTRACT

Neurotrophins, such as brain-derived neurotrophic factor (BDNF), are prominent regulators of neuronal survival, growth and differentiation during development. While trophic factors are viewed as well-understood but not innovative molecules, there are many lines of evidence indicating that BDNF plays an important role in the pathophysiology of many neurodegenerative disorders, depression, anxiety and other psychiatric disorders. In particular, lower levels of BDNF are associated with the aetiology of Alzheimer's and Huntington's diseases. A major challenge is to explain how neurotrophins are able to induce plasticity, improve learning and memory and prevent age-dependent cognitive decline through receptor signalling. This article will review the mechanism of action of neurotrophins and how BDNF/tropomyosin receptor kinase B (TrkB) receptor signaling can dictate trophic responses and change brain plasticity through activity-dependent stimulation. Alternative approaches for modulating BDNF/TrkB signalling to deliver relevant clinical outcomes in neurodegenerative and neuropsychiatric disorders will also be described.


Subject(s)
Nerve Growth Factors/metabolism , Neurodegenerative Diseases/metabolism , Signal Transduction , Animals , Brain/metabolism , Humans , Nerve Growth Factors/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism
18.
Proc Natl Acad Sci U S A ; 116(43): 21343-21345, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31527273
19.
J Neurosci ; 35(34): 11911-20, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311773

ABSTRACT

The p75 neurotrophin receptor (p75(NTR)) is a multifunctional receptor that participates in many critical processes in the nervous system, ranging from apoptosis to synaptic plasticity and morphological events. It is a member of the tumor necrosis factor receptor (TNFR) superfamily, whose members undergo trimeric oligomerization. Interestingly, p75(NTR) interacts with dimeric ligands (i.e., proneurotrophins or mature neurotrophins), but several of the intracellular adaptors that mediate p75(NTR) signaling are trimeric (i.e., TNFR-associated factor 6 or TRAF6). Consequently, the active receptor signaling unit remains uncertain. To identify the functional receptor complex, we evaluated its oligomerization in vitro and in mice brain tissues using a combination of biochemical techniques. We found that the most abundant homotypic arrangement for p75(NTR) is a trimer and that monomers and trimers coexist at the cell surface. Interestingly, trimers are not required for ligand-independent or ligand-dependent p75(NTR) activation in a growth cone retraction functional assay. However, monomers are capable of inducing acute morphological effects in neurons. We propose that p75(NTR) activation is regulated by its oligomerization status and its levels of expression. These results indicate that the oligomeric state of p75(NTR) confers differential responses and offers an explanation for the diverse and contradictory actions of this receptor in the nervous system. SIGNIFICANCE STATEMENT: The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including apoptosis, neuronal processes remodeling, and synaptic plasticity. The goal of our work was to inquire whether oligomers of the receptor are required for function. Here we report that p75(NTR) predominantly assembles as a trimer, similar to other tumor necrosis factor receptors. Interestingly, monomers and trimers coexist at the cell surface, but trimers are not required for p75(NTR) activation in a functional assay. However, monomers are capable of inducing acute morphological effects in neurons. Identification of the oligomerization state of p75(NTR) begins to provide insights to the mechanisms of signal initiation of this noncatalytic receptor, as well as to develop therapeutic interventions to diminish its activity.


Subject(s)
Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/chemistry , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Female , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , PC12 Cells , Rats , Receptors, Nerve Growth Factor/genetics , Stereoisomerism
20.
Proc Natl Acad Sci U S A ; 109(4): 1305-10, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22232675

ABSTRACT

Regulation of the hypothalamic-pituitary-adrenal (HPA) axis is critical for adaptation to environmental changes. The principle regulator of the HPA axis is corticotrophin-releasing hormone (CRH), which is made in the parventricular nucleus and is an important target of negative feedback by glucocorticoids. However, the molecular mechanisms that regulate CRH are not fully understood. Disruption of normal HPA axis activity is a major risk factor of neuropsychiatric disorders in which decreased expression of the glucocorticoid receptor (GR) has been documented. To investigate the role of the GR in CRH neurons, we have targeted the deletion of the GR, specifically in the parventricular nucleus. Impairment of GR function in the parventricular nucleus resulted in an enhancement of CRH expression and an up-regulation of hypothalamic levels of BDNF and disinhibition of the HPA axis. BDNF is a stress and activity-dependent factor involved in many activities modulated by the HPA axis. Significantly, ectopic expression of BDNF in vivo increased CRH, whereas reduced expression of BDNF, or its receptor TrkB, decreased CRH expression and normal HPA functions. We find the differential regulation of CRH relies upon the cAMP response-element binding protein coactivator CRTC2, which serves as a switch for BDNF and glucocorticoids to direct the expression of CRH.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Corticotropin-Releasing Hormone/metabolism , Glucocorticoids/metabolism , Homeostasis/physiology , Hypothalamus/physiology , Receptors, Glucocorticoid/metabolism , Trans-Activators/metabolism , Analysis of Variance , Animals , Chromatin Immunoprecipitation , Hypothalamo-Hypophyseal System/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Mutagenesis, Site-Directed , Neurons/metabolism , Pituitary-Adrenal System/physiology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL