Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Epidemiol ; 191(6): 1040-1049, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35029630

ABSTRACT

Although air pollution is an important risk factor for stroke, few studies have considered the impact of workplace exposure to particulate matter (PM). We examined implications of exposure to PM composed of metalworking fluids (MWFs) for stroke mortality in the United Autoworkers-General Motors cohort. Cox proportional hazards models with age as the timescale were used to estimate the association of cumulative straight, soluble, and synthetic MWF exposure with stroke mortality, controlling for sex, race, plant, calendar year, and hire year. Among 38,553 autoworkers followed during 1941-1995, we identified 114 ischemic stroke deaths and 113 hemorrhagic stroke deaths. Overall stroke mortality risk was increased among workers in the middle exposure category for straight MWF (hazard ratio (HR) = 1.31, 95% confidence interval (CI): 0.87, 1.98) and workers in the highest exposure category for synthetic MWF (HR = 1.94, 95% CI: 1.13, 3.16) compared with workers who had no direct exposure. Ischemic stroke mortality risk was increased among workers in the highest exposure categories for straight MWF (HR = 1.45, 95% CI: 0.83, 2.52) and synthetic MWF (HR = 2.39, 95% CI: 1.39, 4.50). We observed no clear relationship between MWF exposure and hemorrhagic stroke mortality. Our results support a potentially important role for occupational PM exposures in stroke mortality and indicate the need for further studies of PM exposure and stroke in varied occupational settings.


Subject(s)
Hemorrhagic Stroke , Ischemic Stroke , Occupational Diseases , Occupational Exposure , Automobiles , Humans , Metallurgy , Occupational Diseases/etiology , Occupational Exposure/adverse effects , Particulate Matter/adverse effects
2.
Environ Health ; 20(1): 45, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33865403

ABSTRACT

BACKGROUND: Migraine-an episodic disorder characterized by severe headache that can lead to disability-affects over 1 billion people worldwide. Prior studies have found that short-term exposure to fine particulate matter (PM2.5), nitrogen dioxide (NO2), and ozone increases risk of migraine-related emergency department (ED) visits. Our objective was to characterize the association between long-term exposure to sources of harmful emissions and common air pollutants with both migraine headache and, among patients with migraine, headache severity. METHODS: From the Sutter Health electronic health record database, we identified 89,575 prevalent migraine cases between 2014 and 2018 using a migraine probability algorithm (MPA) score and 270,564 frequency-matched controls. Sutter Health delivers care to 3.5 million patients annually in Northern California. Exposures included 2015 annual average block group-level PM2.5 and NO2 concentrations, inverse-distance weighted (IDW) methane emissions from 60 super-emitters located within 10 km of participant residence between 2016 and 2018, and IDW active oil and gas wells in 2015 within 10 km of each participant. We used logistic and negative binomial mixed models to evaluate the association between environmental exposures and (1) migraine case status; and (2) migraine severity (i.e., MPA score > 100, triptan prescriptions, neurology visits, urgent care migraine visits, and ED migraine visits per person-year). Models controlled for age, sex, race/ethnicity, Medicaid use, primary care visits, and block group-level population density and poverty. RESULTS: In adjusted analyses, for each 5 ppb increase in NO2, we observed 2% increased odds of migraine case status (95% CI: 1.00, 1.05) and for each 100,000 kg/hour increase in IDW methane emissions, the odds of case status also increased (OR = 1.04, 95% CI: 1.00, 1.08). We found no association between PM2.5 or oil and gas wells and migraine case status. PM2.5 was linearly associated with neurology visits, migraine-specific urgent care visits, and MPA score > 100, but not triptans or ED visits. NO2 was associated with migraine-specific urgent care and ED visits, but not other severity measures. We observed limited or null associations between continuous measures of methane emissions and proximity to oil and gas wells and migraine severity. CONCLUSIONS: Our findings illustrate the potential role of long-term exposure to multiple ambient air pollutants for prevalent migraine and migraine severity.


Subject(s)
Air Pollutants/analysis , Air Pollution/analysis , Methane/analysis , Migraine Disorders/epidemiology , Nitrogen Dioxide/analysis , Oil and Gas Fields , Particulate Matter/analysis , Adolescent , Adult , Aged , Ambulatory Care/statistics & numerical data , California/epidemiology , Case-Control Studies , Emergency Service, Hospital/statistics & numerical data , Female , Humans , Male , Middle Aged , Prevalence , Severity of Illness Index , Young Adult
3.
Epilepsia ; 57(8): e168-72, 2016 08.
Article in English | MEDLINE | ID: mdl-27344989

ABSTRACT

Vigabatrin is a highly effective antiseizure medication, but its use is limited due to concerns about retinal toxicity. One proposed mechanism for this toxicity is vigabatrin-mediated reduction of taurine. Herein we assess plasma taurine levels in a retrospective cohort of children with epilepsy, including a subset receiving vigabatrin. All children who underwent a plasma amino acid analysis as part of their clinical evaluation between 2006 and 2015 at Stanford Children's Health were included in the analysis. There were no significant differences in plasma taurine levels between children taking vigabatrin (n = 16), children taking other anti-seizure medications, and children not taking any anti-seizure medication (n = 556) (analysis of variance [ANOVA] p = 0.841). There were, however, age-dependent decreases in plasma taurine levels. Multiple linear regression revealed no significant association between vigabatrin use and plasma taurine level (p = 0.87) when controlling for age. These results suggest that children taking vigabatrin maintain normal plasma taurine levels, although they leave unanswered whether taurine supplementation is necessary or sufficient to prevent vigabatrin-associated visual field loss. They also indicate that age should be taken into consideration when evaluating taurine levels in young children.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/blood , Epilepsy/drug therapy , Taurine/blood , Vigabatrin/therapeutic use , Age Factors , Analysis of Variance , Child, Preschool , Cohort Studies , Female , Humans , Male
5.
Acta Neuropathol ; 127(4): 593-603, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24196163

ABSTRACT

Neural tumors often express neurotransmitter receptors as markers of their developmental lineage. Although these receptors have been well characterized in electrophysiological, developmental and pharmacological settings, their importance in the maintenance and progression of brain tumors and, importantly, the effect of their targeting in brain cancers remains obscure. Here, we demonstrate high levels of GABRA5, which encodes the α5-subunit of the GABAA receptor complex, in aggressive MYC-driven, "Group 3" medulloblastomas. We hypothesized that modulation of α5-GABAA receptors alters medulloblastoma cell survival and monitored biological and electrophysiological responses of GABRA5-expressing medulloblastoma cells upon pharmacological targeting of the GABAA receptor. While antagonists, inverse agonists and non-specific positive allosteric modulators had limited effects on medulloblastoma cells, a highly specific and potent α5-GABAA receptor agonist, QHii066, resulted in marked membrane depolarization and a significant decrease in cell survival. This effect was GABRA5 dependent and mediated through the induction of apoptosis as well as accumulation of cells in S and G2 phases of the cell cycle. Chemical genomic profiling of QHii066-treated medulloblastoma cells confirmed inhibition of MYC-related transcriptional activity and revealed an enrichment of HOXA5 target gene expression. siRNA-mediated knockdown of HOXA5 markedly blunted the response of medulloblastoma cells to QHii066. Furthermore, QHii066 sensitized GABRA5 positive medulloblastoma cells to radiation and chemotherapy consistent with the role of HOXA5 in directly regulating p53 expression and inducing apoptosis. Thus, our results provide novel insights into the synthetic lethal nature of α5-GABAA receptor activation in MYC-driven/Group 3 medulloblastomas and propose its targeting as a novel strategy for the management of this highly aggressive tumor.


Subject(s)
Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/physiology , Medulloblastoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Nicotinic/metabolism , Animals , Benzodiazepines/pharmacology , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/pathology , Cisplatin/pharmacology , Colony-Forming Units Assay , GABA Agonists/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Medulloblastoma/pathology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Patch-Clamp Techniques , Receptors, Nicotinic/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays , gamma-Aminobutyric Acid/pharmacology
6.
FASEB J ; 27(3): 1264-74, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23221336

ABSTRACT

The mechanism of release and the role of l-aspartate as a central neurotransmitter are controversial. A vesicular release mechanism for l-aspartate has been difficult to prove, as no vesicular l-aspartate transporter was identified until it was found that sialin could transport l-aspartate and l-glutamate when reconstituted into liposomes. We sought to clarify the release mechanism of l-aspartate and the role of sialin in this process by combining l-aspartate uptake studies in isolated synaptic vesicles with immunocyotchemical investigations of hippocampal slices. We found that radiolabeled l-aspartate was taken up into synaptic vesicles. The vesicular l-aspartate uptake, relative to the l-glutamate uptake, was twice as high in the hippocampus as in the whole brain, the striatum, and the entorhinal and frontal cortices and was not inhibited by l-glutamate. We further show that sialin is not essential for exocytosis of l-aspartate, as there was no difference in ATP-dependent l-aspartate uptake in synaptic vesicles from sialin-knockout and wild-type mice. In addition, expression of sialin in PC12 cells did not result in significant vesicle uptake of l-aspartate, and depolarization-induced depletion of l-aspartate from hippocampal nerve terminals was similar in hippocampal slices from sialin-knockout and wild-type mice. Further, there was no evidence for nonvesicular release of l-aspartate via volume-regulated anion channels or plasma membrane excitatory amino acid transporters. This suggests that l-aspartate is exocytotically released from nerve terminals after vesicular accumulation by a transporter other than sialin.


Subject(s)
Aspartic Acid/metabolism , Brain/metabolism , Exocytosis/physiology , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Organic Anion Transporters/metabolism , Symporters/metabolism , Synaptic Vesicles/metabolism , Adenosine Triphosphate/metabolism , Animals , Male , Mice , Mice, Knockout , PC12 Cells , Rats , Rats, Wistar
7.
Article in English | MEDLINE | ID: mdl-37555559

ABSTRACT

Objective: To assemble and characterize an electronic health record (EHR) dataset for a large cohort of US military Veterans diagnosed with ALS (Amyotrophic Lateral Sclerosis). Methods: An EHR dataset for 19,662 Veterans diagnosed with ALS between January 1, 2000 to December 31, 2020 was compiled from the Veterans Health Administration (VHA) EHR database by a query for ICD9 diagnosis (335.20) or ICD10 diagnosis (G12.21) for Amyotrophic Lateral Sclerosis. Results: The cohort is predominantly male (98.94%) and white (72.37%) with a median age at disease onset of 68 years and median survival from the date of diagnosis of 590 days. With the designation of ALS as a compensable illness in 2009, there was a subsequent increase in the number of Veterans diagnosed per year in the VHA, but no change in median survival. The cohort included a greater-than-expected proportion of individuals whose branch of service at the time of separation was the Army. Conclusions: The composition of the cohort reflects the VHA population who are at greatest risk for ALS. The greater than expected proportion of individuals whose branch of service at the time of separation was the Army suggests the possibility of a branch-specific risk factor for ALS.

8.
Neuron ; 110(19): 3186-3205.e7, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35961319

ABSTRACT

Structural plasticity in the brain often necessitates dramatic remodeling of neuronal processes, with attendant reorganization of the cytoskeleton and membranes. Although cytoskeletal restructuring has been studied extensively, how lipids might orchestrate structural plasticity remains unclear. We show that specific glial cells in Drosophila produce glucocerebrosidase (GBA) to locally catabolize sphingolipids. Sphingolipid accumulation drives lysosomal dysfunction, causing gba1b mutants to harbor protein aggregates that cycle across circadian time and are regulated by neural activity, the circadian clock, and sleep. Although the vast majority of membrane lipids are stable across the day, a specific subset that is highly enriched in sphingolipids cycles daily in a gba1b-dependent fashion. Remarkably, both sphingolipid biosynthesis and degradation are required for the diurnal remodeling of circadian clock neurites, which grow and shrink across the day. Thus, dynamic sphingolipid regulation by glia enables diurnal circuit remodeling and proper circadian behavior.


Subject(s)
Circadian Clocks , Drosophila Proteins , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila/metabolism , Drosophila Proteins/metabolism , Glucosylceramidase , Membrane Lipids , Neuroglia/metabolism , Protein Aggregates , Sphingolipids/metabolism
9.
J Neurosci ; 30(4): 1288-300, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20107056

ABSTRACT

The neurotransmitter glutamate is recycled through an astrocytic-neuronal glutamate-glutamine cycle in which synaptic glutamate is taken up by astrocytes, metabolized to glutamine, and transferred to neurons for conversion back to glutamate and subsequent release. The extent to which neuronal glutamate release is dependent upon this pathway remains unclear. Here we provide electrophysiological and biochemical evidence that in acutely disinhibited rat neocortical slices, robust release of glutamate during sustained epileptiform activity requires that neurons be provided a continuous source of glutamine. We demonstrate that the uptake of glutamine into neurons for synthesis of glutamate destined for synaptic release is not strongly dependent on the system A transporters, but requires another unidentified glutamine transporter or transporters. Finally, we find that the attenuation of network activity through inhibition of neuronal glutamine transport is associated with reduced frequency and amplitude of spontaneous events detected at the single-cell level. These results indicate that availability of glutamine influences neuronal release of glutamate during periods of intense network activity.


Subject(s)
Action Potentials/physiology , Epilepsy/metabolism , Glutamine/metabolism , Neocortex/metabolism , Neural Inhibition/physiology , Neurons/metabolism , Action Potentials/drug effects , Animals , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Epilepsy/physiopathology , Glutamic Acid/metabolism , Neocortex/drug effects , Neocortex/physiopathology , Nerve Net/drug effects , Nerve Net/metabolism , Nerve Net/physiopathology , Neural Inhibition/drug effects , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neurons/drug effects , Organ Culture Techniques , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
10.
J Biol Chem ; 285(25): 19316-23, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20424173

ABSTRACT

Salla disease and infantile sialic acid storage disorder are human diseases caused by loss of function of sialin, a lysosomal transporter that mediates H(+)-coupled symport of acidic sugars N-acetylneuraminic acid and glucuronic acid out of lysosomes. Along with the closely related vesicular glutamate transporters, sialin belongs to the SLC17 transporter family. Despite their critical role in health and disease, these proteins remain poorly understood both structurally and mechanistically. Here, we use substituted cysteine accessibility screening and radiotracer flux assays to evaluate experimentally a computationally generated three-dimensional structure model of sialin. According to this model, sialin consists of 12 transmembrane helices (TMs) with an overall architecture similar to that of the distantly related glycerol 3-phosphate transporter GlpT. We show that TM4 in sialin lines a large aqueous cavity that forms a part of the substrate permeation pathway and demonstrate substrate-induced alterations in accessibility of substituted cysteine residues in TM4. In addition, we demonstrate that one mutant, F179C, has a dramatically different effect on the apparent affinity and transport rate for N-acetylneuraminic acid and glucuronic acid, suggesting that it may be directly involved in substrate recognition and/or translocation. These findings offer a basis for further defining the transport mechanism of sialin and other SLC17 family members.


Subject(s)
Organic Anion Transporters/chemistry , Symporters/chemistry , Amino Acid Sequence , Animals , Cysteine/chemistry , Glucuronic Acid/chemistry , Humans , Lysosomal Storage Diseases/metabolism , Molecular Sequence Data , Mutation , N-Acetylneuraminic Acid/chemistry , Protein Isoforms , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Structure-Activity Relationship
11.
Cureus ; 13(7): e16297, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34381654

ABSTRACT

Critical care management of acute respiratory failure in patients with neuromuscular disease (NMD) such as amyotrophic lateral sclerosis (ALS) is not standardized and is challenging for many critical care specialists. Progressive hypercapnic respiratory failure and ineffective airway clearance are key issues in this patient population. Often at the time of hospital presentation, patients are already supported by home mechanical ventilatory support with noninvasive ventilation (NIV) and an airway clearance regimen. Prognosis is poor once a patient develops acute respiratory failure requiring intubation and invasive mechanical ventilatory support, commonly leading to tracheostomy or palliative-focused care.  We focus on this understudied group of patients with ALS without tracheostomy and incorporate existing data to propose a technical approach to the triage and management of acute respiratory failure, primarily for those who require intubation and mechanical ventilatory support for reversible causes, and also for progression of end-stage disease. Optimizing management in this setting improves both quality and quantity of life. Neuromuscular patients with acute respiratory failure require protocolized and personalized triage and treatment. Here, we describe the technical methods used at our single institution. The triage phase incorporates comprehensive evaluation for new etiologies of hypoxia and hypercapnia, which are not initially presumed to be secondary to progression or end-stage neuromuscular respiratory failure. In select patients, this may involve intubation or advanced adjustments of NIV machines. Next, once the acute etiology(s) is identified and treated, the focus shifts: training and use of mechanical airway clearance to optimize pulmonary function, facilitation of NIV wean or successful extubation to NIV, and transition to a stable regimen for home ventilation. The comprehensive protocol described here incorporates multi-institutional approaches and effectively optimizes acute respiratory failure in patients with neuromuscular pulmonary disease.

12.
J Neurosci ; 29(49): 15355-65, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-20007460

ABSTRACT

Salla disease and infantile sialic acid storage disease are autosomal recessive lysosomal storage disorders caused by mutations in the gene encoding sialin, a membrane protein that transports free sialic acid out of the lysosome after it is cleaved from sialoglycoconjugates undergoing degradation. Accumulation of sialic acid in lysosomes defines these disorders, and the clinical phenotype is characterized by neurodevelopmental defects, including severe CNS hypomyelination. In this study, we used a sialin-deficient mouse to address how loss of sialin leads to the defect in myelination. Behavioral analysis of the sialin(-/-) mouse demonstrates poor coordination, seizures, and premature death. Analysis by histology, electron microscopy, and Western blotting reveals a decrease in myelination of the CNS but normal neuronal cytoarchitecture and normal myelination of the PNS. To investigate potential mechanisms underlying CNS hypomyelination, we studied myelination and oligodendrocyte development in optic nerves. We found reduced numbers of myelinated axons in optic nerves from sialin(-/-) mice, but the myelin that was present appeared grossly normal. Migration and density of oligodendrocyte precursor cells were normal; however, a marked decrease in the number of postmitotic oligodendrocytes and an associated increase in the number of apoptotic cells during the later stages of myelinogenesis were observed. These findings suggest that a defect in maturation of cells in the oligodendrocyte lineage leads to increased apoptosis and underlies the myelination defect associated with sialin loss.


Subject(s)
Brain/growth & development , Brain/physiology , Myelin Sheath/physiology , Organic Anion Transporters/metabolism , Spinal Cord/growth & development , Spinal Cord/physiology , Symporters/metabolism , Animals , Apoptosis/physiology , Axons/pathology , Axons/physiology , Axons/ultrastructure , Brain/pathology , Cell Count , Cell Movement/physiology , Longevity/physiology , Mice , Mice, Knockout , Motor Activity/physiology , Myelin Basic Protein , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neurons/physiology , Neurons/ultrastructure , Oligodendroglia/pathology , Oligodendroglia/physiology , Oligodendroglia/ultrastructure , Optic Nerve/growth & development , Optic Nerve/pathology , Optic Nerve/ultrastructure , Organic Anion Transporters/genetics , Peripheral Nervous System/growth & development , Peripheral Nervous System/pathology , Peripheral Nervous System/physiology , Seizures/metabolism , Seizures/pathology , Spinal Cord/pathology , Stem Cells/pathology , Stem Cells/physiology , Stem Cells/ultrastructure , Symporters/genetics , Transcription Factors/metabolism
13.
J Cell Biol ; 157(3): 349-55, 2002 Apr 29.
Article in English | MEDLINE | ID: mdl-11980913

ABSTRACT

The transfer of glutamine between cells contributes to signaling as well as to metabolism. The recent identification and characterization of the system N and A family of transporters has begun to suggest mechanisms for the directional transfer of glutamine, and should provide ways to test its physiological significance in diverse processes from nitrogen to neurotransmitter release.


Subject(s)
Glutamine/metabolism , Electrophysiology , Forecasting , Signal Transduction , Synaptic Transmission/physiology
14.
Sci Rep ; 9(1): 14020, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31570734

ABSTRACT

Among high-grade brain tumors, glioblastoma is particularly difficult to treat, in part due to its highly infiltrative nature which contributes to the malignant phenotype and high mortality in patients. In order to better understand the signaling pathways underlying glioblastoma invasion, we performed the first large-scale CRISPR-Cas9 loss of function screen specifically designed to identify genes that facilitate cell invasion. We tested 4,574 genes predicted to be involved in trafficking and motility. Using a transwell invasion assay, we discovered 33 genes essential for invasion. Of the 11 genes we selected for secondary testing using a wound healing assay, 6 demonstrated a significant decrease in migration. The strongest regulator of invasion was mitogen-activated protein kinase 4 (MAP4K4). Targeting of MAP4K4 with single guide RNAs or a MAP4K4 inhibitor reduced migration and invasion in vitro. This effect was consistent across three additional patient derived glioblastoma cell lines. Analysis of epithelial-mesenchymal transition markers in U138 cells with lack or inhibition of MAP4K4 demonstrated protein expression consistent with a non-invasive state. Importantly, MAP4K4 inhibition limited migration in a subset of human glioma organotypic slice cultures. Our results identify MAP4K4 as a novel potential therapeutic target to limit glioblastoma invasion.


Subject(s)
Brain Neoplasms/pathology , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/genetics , Glioblastoma/genetics , Humans , Neoplasm Invasiveness/genetics
15.
Neuron ; 95(4): 779-790.e6, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28817799

ABSTRACT

There is significant need to develop physiologically relevant models for investigating human astrocytes in health and disease. Here, we present an approach for generating astrocyte lineage cells in a three-dimensional (3D) cytoarchitecture using human cerebral cortical spheroids (hCSs) derived from pluripotent stem cells. We acutely purified astrocyte-lineage cells from hCSs at varying stages up to 20 months in vitro using immunopanning and cell sorting and performed high-depth bulk and single-cell RNA sequencing to directly compare them to purified primary human brain cells. We found that hCS-derived glia closely resemble primary human fetal astrocytes and that, over time in vitro, they transition from a predominantly fetal to an increasingly mature astrocyte state. Transcriptional changes in astrocytes are accompanied by alterations in phagocytic capacity and effects on neuronal calcium signaling. These findings suggest that hCS-derived astrocytes closely resemble primary human astrocytes and can be used for studying development and modeling disease.


Subject(s)
Astrocytes/physiology , Cerebral Cortex/cytology , Models, Biological , Pluripotent Stem Cells/physiology , Cell Differentiation/physiology , Cells, Cultured , Fetus , Humans , Imaging, Three-Dimensional , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Neuron ; 93(4): 897-913.e7, 2017 Feb 22.
Article in English | MEDLINE | ID: mdl-28231469

ABSTRACT

Mechanisms regulating the surveillance and clearance of synaptic proteins are not well understood. Intriguingly, the loss of the presynaptic active zone proteins Piccolo and Bassoon triggers the loss of synaptic vesicles (SVs) and compromises synaptic integrity. Here we report that the destruction of SVs in boutons lacking Piccolo and Bassoon was associated with the induction of presynaptic autophagy, a process that depended on poly-ubiquitination, but not the E3 ubiquitin ligase Siah1. Surprisingly, gain or loss of function (LOF) of Bassoon alone suppressed or enhanced presynaptic autophagy, respectively, implying a fundamental role for Bassoon in the local regulation of presynaptic autophagy. Mechanistically, Bassoon was found to interact with Atg5, an E3-like ligase essential for autophagy, and to inhibit the induction of autophagy in heterologous cells. Importantly, Atg5 LOF as well as targeting an Atg5-binding peptide derived from Bassoon inhibited presynaptic autophagy in boutons lacking Piccolo and Bassoon, providing insights into the molecular mechanisms regulating presynaptic autophagy.


Subject(s)
Autophagy-Related Protein 5/metabolism , Autophagy/physiology , Synaptic Vesicles/metabolism , Animals , Nerve Tissue Proteins/metabolism , Presynaptic Terminals/metabolism , Rats , Ubiquitination
17.
PLoS One ; 11(12): e0167535, 2016.
Article in English | MEDLINE | ID: mdl-27907191

ABSTRACT

Synaptic vesicles (SVs) fuse with the plasma membrane at a precise location called the presynaptic active zone (AZ). This fusion is coordinated by proteins embedded within a cytoskeletal matrix assembled at the AZ (CAZ). In the present study, we have identified a novel binding partner for the CAZ proteins Piccolo and Bassoon. This interacting protein, Trio, is a member of the Dbl family of guanine nucleotide exchange factors (GEFs) known to regulate the dynamic assembly of actin and growth factor dependent axon guidance and synaptic growth. Trio was found to interact with the C-terminal PBH 9/10 domains of Piccolo and Bassoon via its own N-terminal Spectrin repeats, a domain that is also critical for its localization to the CAZ. Moreover, our data suggest that regions within the C-terminus of Trio negatively regulate its interactions with Piccolo/Bassoon. These findings provide a mechanism for the presynaptic targeting of Trio and support a model in which Piccolo and Bassoon play a role in regulating neurotransmission through interactions with proteins, including Trio, that modulate the dynamic assembly of F-actin during cycles of synaptic vesicle exo- and endocytosis.


Subject(s)
Cytoskeletal Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neuropeptides/genetics , Presynaptic Terminals/metabolism , Protein Serine-Threonine Kinases/genetics , Synaptic Transmission/genetics , Actins/genetics , Actins/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Cytoskeletal Proteins/metabolism , Embryo, Mammalian , Endocytosis , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Neuropeptides/metabolism , Presynaptic Terminals/ultrastructure , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure
18.
J Neurosci ; 23(2): 518-29, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12533612

ABSTRACT

Vesicular transporters regulate the amount and type of neurotransmitter sequestered into synaptic vesicles and, hence, the kind of signal transmitted to postsynaptic neurons. Glutamate is the prominent excitatory neurotransmitter in retina; GABA and glycine are the main inhibitory neurotransmitters. Little is known about the ontogeny of vesicular neurotransmission in retina. We investigated expression of glutamatergic [vesicular glutamate transporter 1 (VGLUT1)] and GABA/glycinergic [vesicular GABA/glycine transporter (VGAT)] vesicular transporters in postnatal retina. VGLUT1 labels glutamatergic synapses. VGLUT1 and synaptic vesicle 2 colocalized to photoreceptor terminals. VGLUT1 colocalized with PKC to rod bipolar terminals and to ON bipolar terminals in metabotropic glutamate receptor 6+/- mice. Developmentally, VGAT expression precedes VGLUT1. In rat and mouse retina, VGAT occurred in the inner retina by postnatal day 1 (P1). In rat retina, VGLUT1 was in the outer retina by P5-P7 and the inner retina by P7. In the mouse retina, VGLUT1 expression was in the outer retina by P3 and the inner retina by P5. Both rat and mouse retina had an adult pattern of VGLUT1 expression by P14. VGLUT1 expression precedes ribbon synapses, which are first observed in the inner retina at P11 (Fisher, 1979) in mouse and P13 (Horsburgh and Sefton, 1987) in rat. The ribbon synapse marker RIBEYE was not detected in inner retina of P5 or P7 rat. Spontaneous EPSCs in mouse ganglion cells were recorded as early as P7. Together, these findings indicate that vesicular GABA and glycine transmission precedes vesicular glutamate transmission in developing rodent retina. Furthermore, vesicular glutamate transmission likely occurs before ribbon synapse formation in the inner retina.


Subject(s)
Carrier Proteins/biosynthesis , Glutamic Acid/metabolism , Glycine/metabolism , Membrane Proteins/biosynthesis , Membrane Transport Proteins , Organic Anion Transporters , Retina/metabolism , Vesicular Transport Proteins , gamma-Aminobutyric Acid/metabolism , Age Factors , Animals , Excitatory Postsynaptic Potentials/physiology , GABA Plasma Membrane Transport Proteins , Heterozygote , In Vitro Techniques , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Tissue Proteins/biosynthesis , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/ultrastructure , Presynaptic Terminals/metabolism , Rats , Rats, Long-Evans , Retina/cytology , Retina/growth & development , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Vesicular Glutamate Transport Protein 1
19.
J Neurosci ; 23(4): 1265-75, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12598615

ABSTRACT

Recent work has identified a lysosomal protein that transports neutral amino acids (LYAAT1). We now show that LYAAT1 mediates H+ cotransport with a stoichiometry of 1 H+/1 amino acid, consistent with a role in the active efflux of amino acids from lysosomes. In neurons, however, LYAAT1 localizes to axonal processes as well as lysosomes. In axons LYAAT1 fails to colocalize with synaptic markers. Rather, axonal LYAAT1 colocalizes with the exocyst, suggesting a role for membranes expressing LYAAT1 in specifying sites for exocytosis. A protease protection assay and measurements of intracellular pH further indicate abundant expression at the plasma membrane, raising the possibility of physiological roles for LYAAT1 on the cell surface as well as in lysosomes.


Subject(s)
Amino Acid Transport Systems, Neutral/analysis , Amino Acid Transport Systems, Neutral/physiology , Amino Acid Transport Systems/analysis , Amino Acid Transport Systems/physiology , Hippocampus/chemistry , Neurons/chemistry , Protons , Amino Acid Sequence , Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems, Neutral/chemistry , Amino Acids/metabolism , Animals , Axons/chemistry , Biological Transport, Active , Cell Membrane/chemistry , Cells, Cultured , Exocytosis , HeLa Cells , Hippocampus/cytology , Humans , Ion Transport , Lysosomes/chemistry , Molecular Sequence Data , Neurons/metabolism , Patch-Clamp Techniques , Rats , Sequence Alignment , Symporters , Xenopus
20.
J Neurosci ; 22(1): 62-72, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756489

ABSTRACT

Astrocytes provide the glutamine required by neurons to synthesize glutamate and GABA. However, the mechanisms involved in glutamine transfer from glia to neurons have remained poorly understood. Recent work has implicated the System N transporter SN1 in the efflux of glutamine from astrocytes and the very closely related System A transporters SA1 and SA2 in glutamine uptake by neurons. To understand how these closely related proteins mediate flux in different directions, we have examined their ionic coupling. In contrast to the electroneutral exchange of H+ for Na+ and neutral amino acid catalyzed by SN1, we now show that SA1 and SA2 do not couple H+ movement to amino acid flux. As a result, SA1 and SA2 are electrogenic and do not mediate flux reversal as readily as SN1. Differences between System N and A transporters in coupling to H+ thus contribute to the delivery of glutamine from glia to neurons. Nonetheless, although they are not transported, H+ inhibit SA1 and SA2 by competing with Na+.


Subject(s)
Amino Acid Transport System A/metabolism , Amino Acid Transport Systems, Neutral , Glutamine/metabolism , Membrane Transport Proteins , Neurons/metabolism , Protons , beta-Alanine/analogs & derivatives , Amino Acid Transport System A/antagonists & inhibitors , Amino Acid Transport System A/genetics , Animals , Binding, Competitive/physiology , Biological Transport/physiology , Carrier Proteins/metabolism , Cells, Cultured , Glutamine/pharmacokinetics , Glutamine/pharmacology , Hippocampus , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Potentials/drug effects , Molecular Sequence Data , Neural Inhibition/physiology , Neurons/cytology , Neurons/drug effects , Organ Specificity , Protein Binding/physiology , Rats , Sodium/metabolism , Transfection , beta-Alanine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL