Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 124
1.
Neurobiol Dis ; 174: 105887, 2022 Nov.
Article En | MEDLINE | ID: mdl-36209950

We have previously reported that the single transmembrane protein Dipeptidyl Peptidase Like 6 (DPP6) impacts neuronal and synaptic development. DPP6-KO mice are impaired in hippocampal-dependent learning and memory and exhibit smaller brain size. Recently, we have described novel structures in hippocampal area CA1 in aging mice, apparently derived from degenerating presynaptic terminals, that are significantly more prevalent in DPP6-KO mice compared to WT mice of the same age and that these structures were observed earlier in development in DPP6-KO mice. These novel structures appear as clusters of large puncta that colocalize NeuN, synaptophysin, and chromogranin A, and also partially label for MAP2, amyloid ß, APP, α-synuclein, and phosphorylated tau, with synapsin-1 and VGluT1 labeling on their periphery. In this current study, using immunofluorescence and electron microscopy, we confirm that both APP and amyloid ß are prevalent in these structures; and we show with immunofluorescence the presence of similar structures in humans with Alzheimer's disease. Here we also found evidence that aging DPP6-KO mutants show additional changes related to Alzheimer's disease. We used in vivo MRI to show reduced size of the DPP6-KO brain and hippocampus. Aging DPP6-KO hippocampi contained fewer total neurons and greater neuron death and had diagnostic biomarkers of Alzheimer's disease present including accumulation of amyloid ß and APP and increase in expression of hyper-phosphorylated tau. The amyloid ß and phosphorylated tau pathologies were associated with neuroinflammation characterized by increases in microglia and astrocytes. And levels of proinflammatory or anti-inflammatory cytokines increased in aging DPP6-KO mice. We finally show that aging DPP6-KO mice display circadian dysfunction, a common symptom of Alzheimer's disease. Together these results indicate that aging DPP6-KO mice show symptoms of enhanced neurodegeneration reminiscent of dementia associated with a novel structure resulting from synapse loss and neuronal death. This study continues our laboratory's work in discerning the function of DPP6 and here provides compelling evidence of a direct role of DPP6 in Alzheimer's disease.


Alzheimer Disease , Humans , Mice , Animals , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Aging/pathology , Hippocampus/metabolism , Synapses/metabolism , Mice, Transgenic , tau Proteins/genetics , tau Proteins/metabolism , Amyloid beta-Protein Precursor/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels/metabolism
2.
iScience ; 25(10): 105072, 2022 Oct 21.
Article En | MEDLINE | ID: mdl-36147950

In the axon terminal, microtubule stability is decreased relative to the axon shaft. The dynamic microtubule plus ends found in the axon terminal have many functions, including serving as a docking site for the Cytoplasmic dynein motor. Here, we report an unexplored function of dynein in microtubule regulation in axon terminals: regulation of microtubule stability. Using a forward genetic screen, we identified a mutant with an abnormal axon terminal structure owing to a loss of function mutation in NudC. We show that, in the axon terminal, NudC is a chaperone for the protein Lis1. Decreased Lis1 in nudc axon terminals causes dynein/dynactin accumulation and increased microtubule stability. Microtubule dynamics can be restored by pharmacologically inhibiting dynein, implicating excess dynein motor function in microtubule stabilization. Together, our data support a model in which local NudC-Lis1 modulation of the dynein motor is critical for the regulation of microtubule stability in the axon terminal.

3.
Neurosci Res ; 182: 25-31, 2022 Sep.
Article En | MEDLINE | ID: mdl-35688289

BAX is a Bcl-2 family protein acting on apoptosis. It also promotes mitochondrial fusion by interacting with the mitochondrial fusion protein Mitofusin (Mfn1 and Mfn2). Neuronal mitochondria are important for the development and modification of dendritic spines, which are subcellular compartments accommodating excitatory synapses in postsynaptic neurons. The abundance of dendritic mitochondria influences dendritic spine development. Mitochondrial fusion is essential for mitochondrial homeostasis. Here, we show that in the hippocampal neuron of BAX knockout mice, mitochondrial fusion is impaired, leading to decreases in mitochondrial length and total mitochondrial mass in dendrites. Notably, BAX knockout mice also have fewer dendritic spines and less cellular Adenosine 5'triphosphate (ATP) in dendrites. The spine and ATP changes are abolished by restoring mitochondria fusion via overexpressing Mfn1 and Mfn2. These findings indicate that BAX-mediated mitochondrial fusion in neurons is crucial for the development of dendritic spines and the maintenance of cellular ATP levels.


Dendritic Spines , Mitochondrial Dynamics , Adenosine Triphosphate , Animals , Dendritic Spines/metabolism , GTP Phosphohydrolases/metabolism , Mice , Mitochondrial Proteins/metabolism , bcl-2-Associated X Protein/metabolism
4.
STAR Protoc ; 3(1): 101154, 2022 03 18.
Article En | MEDLINE | ID: mdl-35169716

Electron microscopy (EM) is considered the gold standard for studying macroautophagy and mitophagy, essential cellular processes for brain health. Here, we present a protocol using EM to analyze autophagosomes and mitophagosomes in the mouse amygdala. We describe the preparation of brain sections, followed by staining and EM imaging. We then detail the steps to identify and analyze autophagosome-like and mitophagosome-like structures. This protocol can be easily adapted to analyze autophagosomes and mitophagosomes in other mouse brain regions. For complete details on the use and execution of this protocol, please refer to Duan et al. (2021).


Autophagosomes , Mitophagy , Animals , Brain/diagnostic imaging , Mice , Microscopy, Electron , Staining and Labeling
5.
Cell Rep ; 38(3): 110264, 2022 01 18.
Article En | MEDLINE | ID: mdl-35045307

The subthreshold voltage-gated transient K+ current (IA) carried by pore-forming Kv4.2 subunits regulates the propagation of synaptic input, dendritic excitability, and synaptic plasticity in CA1 pyramidal neuron dendrites of the hippocampus. We report that the Ca2+ channel subunit Cav2.3 regulates IA in this cell type. We initially identified Cav2.3 as a Kv4.2-interacting protein in a proteomic screen and we confirmed Cav2.3-Kv4.2 complex association using multiple techniques. Functionally, Cav2.3 Ca2+-entry increases Kv4.2-mediated whole-cell current due to an increase in Kv4.2 surface expression. Using pharmacology and Cav2.3 knockout mice, we show that Cav2.3 regulates the dendritic gradient of IA. Furthermore, the loss of Cav2.3 function leads to the enhancement of AMPA receptor-mediated synaptic currents and NMDA receptor-mediated spine Ca2+ influx. These results propose that Cav2.3 and Kv4.2 are integral constituents of an ion channel complex that affects synaptic function in the hippocampus.


Calcium Channels, R-Type/metabolism , Dendrites/metabolism , Hippocampus/metabolism , Shal Potassium Channels/metabolism , Synaptic Transmission/physiology , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Rats , Rats, Sprague-Dawley
6.
Front Neuroanat ; 15: 763427, 2021.
Article En | MEDLINE | ID: mdl-34720893

The post-embedding immunogold (PI) technique for immunolabeling of neuronal tissues utilizing standard thin-section transmission electron microscopy (TEM) continues to be a prime method for understanding the functional localization of key proteins in neuronal function. Its main advantages over other immunolabeling methods for thin-section TEM are (1) fairly accurate and quantifiable localization of proteins in cells; (2) double-labeling of sections using two gold particle sizes; and (3) the ability to perform multiple labeling for different proteins by using adjacent sections. Here we first review in detail a common method for PI of neuronal tissues. This method has two major parts. First, we describe the freeze-substitution embedding method: cryoprotected tissue is frozen in liquid propane via plunge-freezing, and is placed in a freeze-substitution instrument in which the tissue is embedded in Lowicryl at low temperatures. We highlight important aspects of freeze-substitution embedding. Then we outline how thin sections of embedded tissue on grids are labeled with a primary antibody and a secondary gold particle-conjugated antibody, and the particular problems encountered in TEM of PI-labeled sections. In the Discussion, we compare our method both to earlier PI methods and to more recent PI methods used by other laboratories. We also compare TEM immunolabeling using PI vs. various pre-embedding immunolabeling methods, especially relating to neuronal tissue.

7.
Neuron ; 109(23): 3793-3809.e8, 2021 12 01.
Article En | MEDLINE | ID: mdl-34614419

Psychosocial stress is a common risk factor for anxiety disorders. The cellular mechanism for the anxiogenic effect of psychosocial stress is largely unclear. Here, we show that chronic social defeat (CSD) stress in mice causes mitochondrial impairment, which triggers the PINK1-Parkin mitophagy pathway selectively in the amygdala. This mitophagy elevation causes excessive mitochondrial elimination and consequent mitochondrial deficiency. Mitochondrial deficiency in the basolateral amygdalae (BLA) causes weakening of synaptic transmission in the BLA-BNST (bed nucleus of the stria terminalis) anxiolytic pathway and increased anxiety. The CSD-induced increase in anxiety-like behaviors is abolished in Pink1-/- and Park2-/- mice and alleviated by optogenetic activation of the BLA-BNST synapse. This study identifies an unsuspected role of mitophagy in psychogenetic-stress-induced anxiety elevation and reveals that mitochondrial deficiency is sufficient to increase anxiety and underlies the psychosocial-stress-induced anxiety increase. Mitochondria and mitophagy, therefore, can be potentially targeted to ameliorate anxiety.


Basolateral Nuclear Complex , Mitophagy , Animals , Anxiety , Anxiety Disorders , Basolateral Nuclear Complex/metabolism , Mice , Mice, Inbred C57BL , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Front Synaptic Neurosci ; 13: 685052, 2021.
Article En | MEDLINE | ID: mdl-34108873

Invaginating structures are common in the synapses of most animals. However, the details of these invaginating structures remain understudied in part because they are not well resolved in light microscopy and were often misidentified in early electron microscope (EM) studies. Utilizing experimental techniques along with the latest advances in microscopy, such as focused ion beam-scanning EM (FIB-SEM), evidence is gradually building to suggest that the synaptic invaginating structures contribute to synapse development, maintenance, and plasticity. These invaginating structures are most elaborate in synapses mediating rapid integration of signals, such as muscle contraction, mechanoreception, and vision. Here we argue that the synaptic invaginations should be considered in future studies seeking to understand their role in sensory integration and coordination, learning, and memory. We review the various types of invaginating structures in the synapses and discuss their potential functions. We also present several new examples of invaginating structures from a variety of animals including Drosophila and mice, mainly using FIB-SEM, with which we trace the form and arrangement of these structures.

9.
Mol Psychiatry ; 26(9): 4633-4651, 2021 09.
Article En | MEDLINE | ID: mdl-33589740

Mitochondria are cellular ATP generators. They are dynamic structures undergoing fission and fusion. While much is known about the mitochondrial fission machinery, the mechanism of initiating fission and the significance of fission to neurophysiology are largely unclear. Gamma oscillations are synchronized neural activities that impose a great energy challenge to synapses. The cellular mechanism of fueling gamma oscillations has yet to be defined. Here, we show that dysbindin-1, a protein decreased in the brain of individuals with schizophrenia, is required for neural activity-induced fission by promoting Drp1 oligomerization. This process is engaged by gamma-frequency activities and in turn, supports gamma oscillations. Gamma oscillations and novel object recognition are impaired in dysbindin-1 null mice. These defects can be ameliorated by increasing mitochondrial fission. These findings identify a molecular mechanism for activity-induced mitochondrial fission, a role of mitochondrial fission in gamma oscillations, and mitochondrial fission as a potential target for improving cognitive functions.


Mitochondria , Mitochondrial Dynamics , Animals , Dynamins , Dysbindin , Mice , Mice, Knockout , Mitochondrial Proteins , Synapses
10.
FASEB J ; 35(1): e21092, 2021 01.
Article En | MEDLINE | ID: mdl-33378124

Myosin 18Aα is a myosin 2-like protein containing unique N- and C-terminal protein interaction domains that co-assembles with myosin 2. One protein known to bind to myosin 18Aα is ß-Pix, a guanine nucleotide exchange factor (GEF) for Rac1 and Cdc42 that has been shown to promote dendritic spine maturation by activating the assembly of actin and myosin filaments in spines. Here, we show that myosin 18A⍺ concentrates in the spines of cerebellar Purkinje neurons via co-assembly with myosin 2 and through an actin binding site in its N-terminal extension. miRNA-mediated knockdown of myosin 18A⍺ results in a significant defect in spine maturation that is rescued by an RNAi-immune version of myosin 18A⍺. Importantly, ß-Pix co-localizes with myosin 18A⍺ in spines, and its spine localization is lost upon myosin 18A⍺ knockdown or when its myosin 18A⍺ binding site is deleted. Finally, we show that the spines of myosin 18A⍺ knockdown Purkinje neurons contain significantly less F-actin and myosin 2. Together, these data argue that mixed filaments of myosin 2 and myosin 18A⍺ form a complex with ß-Pix in Purkinje neuron spines that promotes spine maturation by enhancing the assembly of actin and myosin filaments downstream of ß-Pix's GEF activity.


Dendritic Spines/metabolism , Myosins/metabolism , Purkinje Cells/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Dendritic Spines/genetics , Gene Deletion , Mice , Myosin Type II/genetics , Myosin Type II/metabolism , Myosins/genetics , Rho Guanine Nucleotide Exchange Factors/genetics
11.
J Neurosci ; 41(6): 1174-1190, 2021 02 10.
Article En | MEDLINE | ID: mdl-33303681

The BAD-BAX-caspase-3 cascade is a canonical apoptosis pathway. Macroautophagy ("autophagy" hereinafter) is a process by which organelles and aggregated proteins are delivered to lysosomes for degradation. Here, we report a new function of the BAD-BAX-caspase-3 cascade and autophagy in the control of synaptic vesicle pools. We found that, in hippocampal neurons of male mice, the BAD-BAX-caspase-3 pathway regulates autophagy, which in turn limits the size of synaptic vesicle pools and influences the kinetics of activity-induced depletion and recovery of synaptic vesicle pools. Moreover, the caspase-autophagy pathway is engaged by fear conditioning to facilitate associative fear learning and memory. This work identifies a new mechanism for controlling synaptic vesicle pools, and a novel, nonapoptotic, presynaptic function of the BAD-BAX-caspase-3 cascade.SIGNIFICANCE STATEMENT Despite the importance of synaptic vesicles for neurons, little is known about how the size of synaptic vesicle pools is maintained under basal conditions and regulated by neural activity. This study identifies a new mechanism for the control of synaptic vesicle pools, and a new, nonapoptotic function of the BAD-BAX-caspase-3 pathway in presynaptic terminals. Additionally, it indicates that autophagy is not only a homeostatic mechanism to maintain the integrity of cells and tissues, but also a process engaged by neural activity to regulate synaptic vesicle pools for optimal synaptic responses, learning, and memory.


Autophagy/physiology , Caspase 3/deficiency , Signal Transduction/physiology , Synaptic Vesicles/metabolism , bcl-2-Associated X Protein/deficiency , bcl-Associated Death Protein/deficiency , Animals , Caspase 3/genetics , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Molecular Imaging/methods , Organ Culture Techniques , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure , bcl-2-Associated X Protein/genetics , bcl-Associated Death Protein/genetics
12.
Acta Neuropathol Commun ; 8(1): 197, 2020 11 23.
Article En | MEDLINE | ID: mdl-33225987

In addition to its role as an auxiliary subunit of A-type voltage-gated K+ channels, we have previously reported that the single transmembrane protein Dipeptidyl Peptidase Like 6 (DPP6) impacts neuronal and synaptic development. DPP6-KO mice are impaired in hippocampal-dependent learning and memory and exhibit smaller brain size. Using immunofluorescence and electron microscopy, we report here a novel structure in hippocampal area CA1 that was significantly more prevalent in aging DPP6-KO mice compared to WT mice of the same age and that these structures were observed earlier in development in DPP6-KO mice. These novel structures appeared as clusters of large puncta that colocalized NeuN, synaptophysin, and chromogranin A. They also partially labeled for MAP2, and with synapsin-1 and VGluT1 labeling on their periphery. Electron microscopy revealed that these structures are abnormal, enlarged presynaptic swellings filled with mainly fibrous material with occasional peripheral, presynaptic active zones forming synapses. Immunofluorescence imaging then showed that a number of markers for aging and especially Alzheimer's disease were found as higher levels in these novel structures in aging DPP6-KO mice compared to WT. Together these results indicate that aging DPP6-KO mice have increased numbers of novel, abnormal presynaptic structures associated with several markers of Alzheimer's disease.


Aging/pathology , CA1 Region, Hippocampal/ultrastructure , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Presynaptic Terminals/ultrastructure , Alzheimer Disease , Animals , Chromogranin A/metabolism , DNA-Binding Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Synapsins/metabolism , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
13.
iScience ; 23(9): 101514, 2020 Sep 25.
Article En | MEDLINE | ID: mdl-32942173

Mitochondrial function relies on multiple quality control mechanisms, including the release of mitochondrial vesicles. To investigate the ultrastructure and prevalence of mitochondrial membranous protrusions (and, by extension, vesicles) in neurons, we surveyed mitochondria in rat and planarian brains using transmission electron microscopy (EM). We observed that mitochondrial protrusions mostly extend from the outer membrane. Leveraging available 3D EM datasets of the brain, we further analyzed mitochondrial protrusions in neurons of mouse and Drosophila brains, identifying high-resolution spatial views of these protrusions. To assess whether the abundance of mitochondrial protrusions and mitochondria-derived vesicles respond to cellular stress, we examined neurons expressing fluorescently tagged mitochondrial markers using confocal microscopy with Airyscan and found increased numbers of mitochondrial protrusions and vesicles with mild stress. Future studies using improved spatial resolution with added temporal information may further define the functional implications of mitochondrial protrusions and vesicles in neurons.

14.
J Clin Invest ; 130(5): 2657-2672, 2020 05 01.
Article En | MEDLINE | ID: mdl-32027617

Hair cells, the mechanosensory receptors of the inner ear, are responsible for hearing and balance. Hair cell death and consequent hearing loss are common results of treatment with ototoxic drugs, including the widely used aminoglycoside antibiotics. Induction of heat shock proteins (HSPs) confers protection against aminoglycoside-induced hair cell death via paracrine signaling that requires extracellular heat shock 70-kDa protein (HSP70). We investigated the mechanisms underlying this non-cell-autonomous protective signaling in the inner ear. In response to heat stress, inner ear tissue releases exosomes that carry HSP70 in addition to canonical exosome markers and other proteins. Isolated exosomes from heat-shocked utricles were sufficient to improve survival of hair cells exposed to the aminoglycoside antibiotic neomycin, whereas inhibition or depletion of exosomes from the extracellular environment abolished the protective effect of heat shock. Hair cell-specific expression of the known HSP70 receptor TLR4 was required for the protective effect of exosomes, and exosomal HSP70 interacted with TLR4 on hair cells. Our results indicate that exosomes are a previously undescribed mechanism of intercellular communication in the inner ear that can mediate nonautonomous hair cell survival. Exosomes may hold potential as nanocarriers for delivery of therapeutics against hearing loss.


Exosomes/metabolism , Hair Cells, Auditory/metabolism , Animals , Anti-Bacterial Agents/toxicity , Cell Communication/drug effects , Cell Communication/physiology , Cell Survival/drug effects , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/pathology , Heat-Shock Response/physiology , In Vitro Techniques , Mice , Mice, Inbred CBA , Mice, Knockout , Models, Biological , Neomycin/toxicity , Ototoxicity/genetics , Ototoxicity/metabolism , Ototoxicity/pathology , Pregnancy , Toll-Like Receptor 4/metabolism , Up-Regulation
15.
Elife ; 82019 10 14.
Article En | MEDLINE | ID: mdl-31609202

Sensory hair cells in the ear utilize specialized ribbon synapses. These synapses are defined by electron-dense presynaptic structures called ribbons, composed primarily of the structural protein Ribeye. Previous work has shown that voltage-gated influx of Ca2+ through CaV1.3 channels is critical for hair-cell synapse function and can impede ribbon formation. We show that in mature zebrafish hair cells, evoked presynaptic-Ca2+ influx through CaV1.3 channels initiates mitochondrial-Ca2+ (mito-Ca2+) uptake adjacent to ribbons. Block of mito-Ca2+ uptake in mature cells depresses presynaptic-Ca2+ influx and impacts synapse integrity. In developing zebrafish hair cells, mito-Ca2+ uptake coincides with spontaneous rises in presynaptic-Ca2+ influx. Spontaneous mito-Ca2+ loading lowers cellular NAD+/NADH redox and downregulates ribbon size. Direct application of NAD+ or NADH increases or decreases ribbon size respectively, possibly acting through the NAD(H)-binding domain on Ribeye. Our results present a mechanism where presynaptic- and mito-Ca2+ couple to confer proper presynaptic function and formation.


Calcium Channels, L-Type/metabolism , Calcium/metabolism , Evoked Potentials, Auditory/physiology , Eye Proteins/metabolism , Hair Cells, Auditory/metabolism , Mitochondria/metabolism , Synapses/metabolism , Zebrafish Proteins/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Animals, Genetically Modified , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Signaling , Cell Size , Embryo, Nonmammalian , Eye Proteins/chemistry , Eye Proteins/genetics , Gene Expression , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Isradipine/pharmacology , Mitochondria/drug effects , Mitochondria/ultrastructure , NAD/metabolism , Oxidation-Reduction , Protein Binding , Protein Interaction Domains and Motifs , Ruthenium Compounds/pharmacology , Synapses/drug effects , Synapses/ultrastructure , Synaptic Transmission , Zebrafish , Zebrafish Proteins/agonists , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
16.
Science ; 366(6462): 246-250, 2019 10 11.
Article En | MEDLINE | ID: mdl-31601770

The function and pharmacology of γ-aminobutyric acid type A receptors (GABAARs) are of great physiological and clinical importance and have long been thought to be determined by the channel pore-forming subunits. We discovered that Shisa7, a single-passing transmembrane protein, localizes at GABAergic inhibitory synapses and interacts with GABAARs. Shisa7 controls receptor abundance at synapses and speeds up the channel deactivation kinetics. Shisa7 also potently enhances the action of diazepam, a classic benzodiazepine, on GABAARs. Genetic deletion of Shisa7 selectively impairs GABAergic transmission and diminishes the effects of diazepam in mice. Our data indicate that Shisa7 regulates GABAAR trafficking, function, and pharmacology and reveal a previously unknown molecular interaction that modulates benzodiazepine action in the brain.


CA1 Region, Hippocampal/physiology , Diazepam/pharmacology , GABA Modulators/pharmacology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pyramidal Cells/physiology , Receptors, GABA-A/metabolism , Synaptic Transmission , Animals , Behavior, Animal/drug effects , Cell Membrane/metabolism , Diazepam/administration & dosage , GABA Modulators/administration & dosage , HEK293 Cells , Humans , Inhibitory Postsynaptic Potentials , Interneurons/physiology , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs , Synapses/physiology
17.
Biol Open ; 8(8)2019 Aug 01.
Article En | MEDLINE | ID: mdl-31362947

Serial-section electron microscopy such as FIB-SEM (focused ion beam scanning electron microscopy) has become an important tool for neuroscientists to trace the trajectories and global architecture of neural circuits in the brain, as well as to visualize the 3D ultrastructure of cellular organelles in neurons. In this study, we examined 3D features of mitochondria in electron microscope images generated from serial sections of four regions of mouse brains: nucleus accumbens (NA), hippocampal CA1, somatosensory cortex and dorsal cochlear nucleus (DCN). We compared mitochondria in the presynaptic terminals to those in the postsynaptic/dendritic compartments, and we focused on the shape and size of mitochondria. A common feature of mitochondria among the four brain regions is that presynaptic mitochondria generally are small and short, and most of them do not extend beyond presynaptic terminals. In contrast, the majority of postsynaptic/dendritic mitochondria are large and many of them spread through significant portions of the dendrites. Comparing among the brain areas, the cerebral cortex and DCN have even larger postsynaptic/dendritic mitochondria than the NA and CA1. Our analysis reveals that mitochondria in neurons are differentially sized and arranged according to their subcellular locations, suggesting a spatial organizing principle of mitochondria at the synapse.

18.
JCI Insight ; 4(12)2019 06 20.
Article En | MEDLINE | ID: mdl-31217345

TRIOBP remodels the cytoskeleton by forming unusually dense F-actin bundles and is implicated in human cancer, schizophrenia, and deafness. Mutations ablating human and mouse TRIOBP-4 and TRIOBP-5 isoforms are associated with profound deafness, as inner ear mechanosensory hair cells degenerate after stereocilia rootlets fail to develop. However, the mechanisms regulating formation of stereocilia rootlets by each TRIOBP isoform remain unknown. Using 3 new Triobp mouse models, we report that TRIOBP-5 is essential for thickening bundles of F-actin in rootlets, establishing their mature dimensions and for stiffening supporting cells of the auditory sensory epithelium. The coiled-coil domains of this isoform are required for reinforcement and maintenance of stereocilia rootlets. A loss of TRIOBP-5 in mouse results in dysmorphic rootlets that are abnormally thin in the cuticular plate but have increased widths and lengths within stereocilia cores, and causes progressive deafness recapitulating the human phenotype. Our study extends the current understanding of TRIOBP isoform-specific functions necessary for life-long hearing, with implications for insight into other TRIOBPopathies.


Hearing/physiology , Microfilament Proteins/physiology , Stereocilia/physiology , Actins/physiology , Animals , Deafness/etiology , Mice , Mice, Knockout , Microfilament Proteins/chemistry , Microfilament Proteins/deficiency , Protein Isoforms/physiology , Stereocilia/ultrastructure
19.
J Biol Chem ; 294(30): 11498-11512, 2019 07 26.
Article En | MEDLINE | ID: mdl-31177092

Neurolastin is a dynamin family GTPase that also contains a RING domain and exhibits both GTPase and E3 ligase activities. It is specifically expressed in the brain and is important for synaptic transmission, as neurolastin knockout animals have fewer dendritic spines and exhibit a reduction in functional synapses. Our initial study of neurolastin revealed that it is membrane-associated and partially co-localizes with endosomes. Using various biochemical and cell-culture approaches, we now show that neurolastin also localizes to mitochondria in HeLa cells, cultured neurons, and brain tissue. We found that the mitochondrial localization of neurolastin depends upon an N-terminal mitochondrial targeting sequence and that neurolastin is imported into the mitochondrial intermembrane space. Although neurolastin was only partially mitochondrially localized at steady state, it displayed increased translocation to mitochondria in response to neuronal stress and mitochondrial fragmentation. Interestingly, inactivation or deletion of neurolastin's RING domain also increased its mitochondrial localization. Using EM, we observed that neurolastin knockout animals have smaller but more numerous mitochondria in cerebellar Purkinje neurons, indicating that neurolastin regulates mitochondrial morphology. We conclude that the brain-specific dynamin GTPase neurolastin exhibits stress-responsive localization to mitochondria and is required for proper mitochondrial morphology.


Dynamins/metabolism , Mitochondria/metabolism , Purkinje Cells/metabolism , Animals , Cells, Cultured , Dynamins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Mutation , Protein Transport
20.
Neuromolecular Med ; 21(3): 314-321, 2019 09.
Article En | MEDLINE | ID: mdl-31177362

Sideroflexin 1 (Sfxn1) is a mitochondrial serine transporter involved in one-carbon metabolism in blood and cancer cell lines. The expression of other Sfxn homologs varies across tissues implying that each homolog may have tissue-specific functions. RNA databases suggest that among the Sfxns, Sfxn3 may have a specific function in the brain. Here, we systematically analyzed the level, cellular distribution, and subcellular localization of Sfxn3 protein in the developing and adult rodent brain. We found that, in the cortex and hippocampus, Sfxn3 protein level is low at birth but increases during development and remains at a high level in the mature brains. Similarly, in cultured hippocampal neurons, Sfxn3 protein level is low in young neurons but increases as neurons mature. Sfxn3 protein level is much higher in neurons than in astrocytes. Within neurons, Sfxn3 localizes to mitochondria in all major neuronal compartments. Our results establish that Sfxn3 is a mitochondrial protein enriched in neurons wherein it is developmentally expressed. These findings provide a foundation for future research aimed at understanding the functions of Sfxn3 and one-carbon metabolism in neurons.


Brain Chemistry , Cation Transport Proteins/physiology , Mitochondrial Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Animals , Astrocytes/metabolism , Brain/cytology , Brain/growth & development , Cation Transport Proteins/analysis , Cells, Cultured , Cerebral Cortex/chemistry , Cerebral Cortex/growth & development , Female , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/growth & development , Male , Mice, Inbred C57BL , Microscopy, Fluorescence , Microscopy, Immunoelectron , Rats , Subcellular Fractions/chemistry
...