Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 102021 06 30.
Article in English | MEDLINE | ID: mdl-34190042

ABSTRACT

The spatiotemporal distribution of mitochondria is crucial for precise ATP provision and calcium buffering required to support neuronal signaling. Fast-spiking GABAergic interneurons expressing parvalbumin (PV+) have a high mitochondrial content reflecting their large energy utilization. The importance for correct trafficking and precise mitochondrial positioning remains poorly elucidated in inhibitory neurons. Miro1 is a Ca²+-sensing adaptor protein that links mitochondria to the trafficking apparatus, for their microtubule-dependent transport along axons and dendrites, in order to meet the metabolic and Ca2+-buffering requirements of the cell. Here, we explore the role of Miro1 in PV+ interneurons and how changes in mitochondrial trafficking could alter network activity in the mouse brain. By employing live and fixed imaging, we found that the impairments in Miro1-directed trafficking in PV+ interneurons altered their mitochondrial distribution and axonal arborization, while PV+ interneuron-mediated inhibition remained intact. These changes were accompanied by an increase in the ex vivo hippocampal γ-oscillation (30-80 Hz) frequency and promoted anxiolysis. Our findings show that precise regulation of mitochondrial dynamics in PV+ interneurons is crucial for proper neuronal signaling and network synchronization.


Subject(s)
Interneurons/physiology , Parvalbumins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Animals, Newborn , Behavior, Animal , Female , Genotype , Hippocampus , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/physiology , Parvalbumins/genetics , rho GTP-Binding Proteins/genetics
2.
Cell Rep ; 26(8): 2037-2051.e6, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30784587

ABSTRACT

Altered excitatory/inhibitory (E/I) balance is implicated in neuropsychiatric and neurodevelopmental disorders, but the underlying genetic etiology remains poorly understood. Copy number variations in CYFIP1 are associated with autism, schizophrenia, and intellectual disability, but its role in regulating synaptic inhibition or E/I balance remains unclear. We show that CYFIP1, and the paralog CYFIP2, are enriched at inhibitory postsynaptic sites. While CYFIP1 or CYFIP2 upregulation increases excitatory synapse number and the frequency of miniature excitatory postsynaptic currents (mEPSCs), it has the opposite effect at inhibitory synapses, decreasing their size and the amplitude of miniature inhibitory postsynaptic currents (mIPSCs). Contrary to CYFIP1 upregulation, its loss in vivo, upon conditional knockout in neocortical principal cells, increases expression of postsynaptic GABAA receptor ß2/3-subunits and neuroligin 3, enhancing synaptic inhibition. Thus, CYFIP1 dosage can bi-directionally impact inhibitory synaptic structure and function, potentially leading to altered E/I balance and circuit dysfunction in CYFIP1-associated neurological disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autistic Disorder/genetics , Brain/physiology , Excitatory Postsynaptic Potentials , Inhibitory Postsynaptic Potentials , Schizophrenia/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/cytology , Brain/metabolism , COS Cells , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Chlorocebus aethiops , Female , Gene Deletion , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Miniature Postsynaptic Potentials , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Synapses/metabolism , Synapses/physiology
3.
Nanoscale ; 10(21): 10241-10249, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29790493

ABSTRACT

Neurons communicate with each other through synapses, which show enrichment for specialized receptors. Although many studies have explored spatial enrichment and diffusion of these receptors in dissociated neurons using single particle tracking, much less is known about their dynamic properties at synapses in complex tissue like brain slices. Here we report the use of smaller and highly specific quantum dots conjugated with a recombinant single domain antibody fragment (VHH fragment) against green fluorescent protein to provide information on diffusion of adhesion molecules at the growth cone and neurotransmitter receptors at synapses. Our data reveals that QD-nanobodies can measure neurotransmitter receptor dynamics at both excitatory and inhibitory synapses in primary neuronal cultures as well as in ex vivo rat brain slices. We also demonstrate that this approach can be applied to tagging multiple proteins to simultaneously monitor their behavior. Thus, we provide a strategy for multiplex imaging of tagged membrane proteins to study their clustering, diffusion and transport both in vitro as well as in native tissue environments such as brain slices.


Subject(s)
Cell Adhesion Molecules/physiology , Neurons/physiology , Quantum Dots , Single-Domain Antibodies/chemistry , Synapses/physiology , Animals , Brain/diagnostic imaging , Diffusion , Green Fluorescent Proteins/chemistry , HeLa Cells , Hippocampus/cytology , Humans , Primary Cell Culture , Rats
4.
EMBO Rep ; 18(2): 231-240, 2017 02.
Article in English | MEDLINE | ID: mdl-28039205

ABSTRACT

Mitochondrial trafficking is influenced by neuronal activity, but it remains unclear how mitochondrial positioning influences neuronal transmission and plasticity. Here, we use live cell imaging with the genetically encoded presynaptically targeted Ca2+ indicator, SyGCaMP5, to address whether presynaptic Ca2+ responses are altered by mitochondria in synaptic terminals. We find that presynaptic Ca2+ signals, as well as neurotransmitter release, are significantly decreased in terminals containing mitochondria. Moreover, the localisation of mitochondria at presynaptic sites can be altered during long-term activity changes, dependent on the Ca2+-sensing function of the mitochondrial trafficking protein, Miro1. In addition, we find that Miro1-mediated activity-dependent synaptic repositioning of mitochondria allows neurons to homeostatically alter the strength of presynaptic Ca2+ signals in response to prolonged changes in neuronal activity. Our results support a model in which mitochondria are recruited to presynaptic terminals during periods of raised neuronal activity and are involved in rescaling synaptic signals during homeostatic plasticity.


Subject(s)
Calcium Signaling , Homeostasis , Mitochondria/metabolism , Neuronal Plasticity , Presynaptic Terminals/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Gene Expression , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
5.
Cell Rep ; 17(2): 317-327, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705781

ABSTRACT

Correct mitochondrial distribution is critical for satisfying local energy demands and calcium buffering requirements and supporting key cellular processes. The mitochondrially targeted proteins Miro1 and Miro2 are important components of the mitochondrial transport machinery, but their specific roles in neuronal development, maintenance, and survival remain poorly understood. Using mouse knockout strategies, we demonstrate that Miro1, as opposed to Miro2, is the primary regulator of mitochondrial transport in both axons and dendrites. Miro1 deletion leads to depletion of mitochondria from distal dendrites but not axons, accompanied by a marked reduction in dendritic complexity. Disrupting postnatal mitochondrial distribution in vivo by deleting Miro1 in mature neurons causes a progressive loss of distal dendrites and compromises neuronal survival. Thus, the local availability of mitochondrial mass is critical for generating and sustaining dendritic arbors, and disruption of mitochondrial distribution in mature neurons is associated with neurodegeneration.


Subject(s)
Dendrites/genetics , Mitochondrial Proteins/genetics , Nerve Degeneration/genetics , Neurogenesis/genetics , rho GTP-Binding Proteins/genetics , Animals , Axons/metabolism , Axons/pathology , Dendrites/metabolism , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology
6.
J Neurosci ; 35(48): 15996-6011, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26631479

ABSTRACT

It is fast emerging that maintaining mitochondrial function is important for regulating astrocyte function, although the specific mechanisms that govern astrocyte mitochondrial trafficking and positioning remain poorly understood. The mitochondrial Rho-GTPase 1 protein (Miro1) regulates mitochondrial trafficking and detachment from the microtubule transport network to control activity-dependent mitochondrial positioning in neurons. However, whether Miro proteins are important for regulating signaling-dependent mitochondrial dynamics in astrocytic processes remains unclear. Using live-cell confocal microscopy of rat organotypic hippocampal slices, we find that enhancing neuronal activity induces transient mitochondrial remodeling in astrocytes, with a concomitant, transient reduction in mitochondrial trafficking, mediated by elevations in intracellular Ca(2+). Stimulating neuronal activity also induced mitochondrial confinement within astrocytic processes in close proximity to synapses. Furthermore, we show that the Ca(2+)-sensing EF-hand domains of Miro1 are important for regulating mitochondrial trafficking in astrocytes and required for activity-driven mitochondrial confinement near synapses. Additionally, activity-dependent mitochondrial positioning by Miro1 reciprocally regulates the levels of intracellular Ca(2+) in astrocytic processes. Thus, the regulation of intracellular Ca(2+) signaling, dependent on Miro1-mediated mitochondrial positioning, could have important consequences for astrocyte Ca(2+) wave propagation, gliotransmission, and ultimately neuronal function.


Subject(s)
Astrocytes/ultrastructure , Calcium Signaling/physiology , Intracellular Space/metabolism , Mitochondria/physiology , Mitochondrial Proteins/metabolism , Synapses/physiology , rho GTP-Binding Proteins/metabolism , Animals , Animals, Newborn , Cells, Cultured , Dependovirus/genetics , Embryo, Mammalian , Excitatory Amino Acid Agents/pharmacology , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/pharmacology , Hippocampus/cytology , In Vitro Techniques , Intracellular Space/genetics , Male , Mitochondrial Proteins/genetics , Neurons/physiology , Organ Culture Techniques , Protein Transport/drug effects , Protein Transport/genetics , Rats , Rats, Sprague-Dawley , Vesicular Glutamate Transport Protein 1/metabolism , rho GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...