Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Science ; 384(6694): 438-446, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38662831

ABSTRACT

Liver mitochondria play a central role in metabolic adaptations to changing nutritional states, yet their dynamic regulation upon anticipated changes in nutrient availability has remained unaddressed. Here, we found that sensory food perception rapidly induced mitochondrial fragmentation in the liver through protein kinase B/AKT (AKT)-dependent phosphorylation of serine 131 of the mitochondrial fission factor (MFFS131). This response was mediated by activation of hypothalamic pro-opiomelanocortin (POMC)-expressing neurons. A nonphosphorylatable MFFS131G knock-in mutation abrogated AKT-induced mitochondrial fragmentation in vitro. In vivo, MFFS131G knock-in mice displayed altered liver mitochondrial dynamics and impaired insulin-stimulated suppression of hepatic glucose production. Thus, rapid activation of a hypothalamus-liver axis can adapt mitochondrial function to anticipated changes of nutritional state in control of hepatic glucose metabolism.


Subject(s)
Food , Gluconeogenesis , Glucose , Liver , Membrane Proteins , Mitochondria, Liver , Mitochondrial Dynamics , Mitochondrial Proteins , Perception , Animals , Male , Mice , Gene Knock-In Techniques , Glucose/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Liver/metabolism , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Neurons/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Transgenic
2.
EMBO Rep ; 24(9): e56766, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37469276

ABSTRACT

During mitotic entry of vertebrate cells, nuclear pore complexes (NPCs) are rapidly disintegrated. NPC disassembly is initiated by hyperphosphorylation of linker nucleoporins (Nups), which leads to the dissociation of FG repeat Nups and relaxation of the nuclear permeability barrier. However, less is known about disintegration of the huge nuclear and cytoplasmic rings, which are formed by annular assemblies of Y-complexes that are dissociated from NPCs as intact units. Surprisingly, we observe that Y-complex Nups display slower dissociation kinetics compared with other Nups during in vitro NPC disassembly, indicating a mechanistic difference in the disintegration of Y-based rings. Intriguingly, biochemical experiments reveal that a fraction of Y-complexes remains associated with mitotic ER membranes, supporting recent microscopic observations. Visualization of mitotic Y-complexes by super-resolution microscopy demonstrates that they form two classes of higher order assemblies: large clusters at kinetochores and small, focal ER-associated assemblies. These, however, lack features qualifying them as persisting ring-shaped subassemblies previously proposed to serve as structural templates for NPC reassembly during mitotic exit, which helps to refine current models of nuclear reassembly.


Subject(s)
Microscopy , Mitosis , Nuclear Pore , Cell Nucleus , Nuclear Pore Complex Proteins/genetics
3.
Nature ; 593(7859): 435-439, 2021 05.
Article in English | MEDLINE | ID: mdl-33953403

ABSTRACT

Mitochondrial fission is a highly regulated process that, when disrupted, can alter metabolism, proliferation and apoptosis1-3. Dysregulation has been linked to neurodegeneration3,4, cardiovascular disease3 and cancer5. Key components of the fission machinery include the endoplasmic reticulum6 and actin7, which initiate constriction before dynamin-related protein 1 (DRP1)8 binds to the outer mitochondrial membrane via adaptor proteins9-11, to drive scission12. In the mitochondrial life cycle, fission enables both biogenesis of new mitochondria and clearance of dysfunctional mitochondria through mitophagy1,13. Current models of fission regulation cannot explain how those dual fates are decided. However, uncovering fate determinants is challenging, as fission is unpredictable, and mitochondrial morphology is heterogeneous, with ultrastructural features that are below the diffraction limit. Here, we used live-cell structured illumination microscopy to capture mitochondrial dynamics. By analysing hundreds of fissions in African green monkey Cos-7 cells and mouse cardiomyocytes, we discovered two functionally and mechanistically distinct types of fission. Division at the periphery enables damaged material to be shed into smaller mitochondria destined for mitophagy, whereas division at the midzone leads to the proliferation of mitochondria. Both types are mediated by DRP1, but endoplasmic reticulum- and actin-mediated pre-constriction and the adaptor MFF govern only midzone fission. Peripheral fission is preceded by lysosomal contact and is regulated by the mitochondrial outer membrane protein FIS1. These distinct molecular mechanisms explain how cells independently regulate fission, leading to distinct mitochondrial fates.


Subject(s)
Mitochondria/metabolism , Mitochondrial Dynamics , Mitophagy , Actins , Animals , COS Cells , Cell Survival , Cells, Cultured , Chlorocebus aethiops , DNA, Mitochondrial/analysis , DNA, Mitochondrial/metabolism , Dynamins , Endoplasmic Reticulum , Humans , Lysosomes , Membrane Proteins , Mice , Mitochondria/genetics , Mitochondrial Proteins
4.
Cell Rep ; 35(2): 108947, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852852

ABSTRACT

During mitochondrial fission, key molecular and cellular factors assemble on the outer mitochondrial membrane, where they coordinate to generate constriction. Constriction sites can eventually divide or reverse upon disassembly of the machinery. However, a role for membrane tension in mitochondrial fission, although speculated, has remained undefined. We capture the dynamics of constricting mitochondria in mammalian cells using live-cell structured illumination microscopy (SIM). By analyzing the diameters of tubules that emerge from mitochondria and implementing a fluorescence lifetime-based mitochondrial membrane tension sensor, we discover that mitochondria are indeed under tension. Under perturbations that reduce mitochondrial tension, constrictions initiate at the same rate, but are less likely to divide. We propose a model based on our estimates of mitochondrial membrane tension and bending energy in living cells which accounts for the observed probability distribution for mitochondrial constrictions to divide.


Subject(s)
Cytoskeleton/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Membranes/metabolism , Animals , Biomechanical Phenomena , COS Cells , Chlorocebus aethiops , Cytoskeleton/ultrastructure , Dynamins/genetics , Dynamins/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/ultrastructure , Surface Tension , Transfection , Transgenes , Red Fluorescent Protein
5.
J Cell Biol ; 220(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33538762

ABSTRACT

Neuronal remodeling and myelination are two fundamental processes during neurodevelopment. How they influence each other remains largely unknown, even though their coordinated execution is critical for circuit function and often disrupted in neuropsychiatric disorders. It is unclear whether myelination stabilizes axon branches during remodeling or whether ongoing remodeling delays myelination. By modulating synaptic transmission, cytoskeletal dynamics, and axonal transport in mouse motor axons, we show that local axon remodeling delays myelination onset and node formation. Conversely, glial differentiation does not determine the outcome of axon remodeling. Delayed myelination is not due to a limited supply of structural components of the axon-glial unit but rather is triggered by increased transport of signaling factors that initiate myelination, such as neuregulin. Further, transport of promyelinating signals is regulated via local cytoskeletal maturation related to activity-dependent competition. Our study reveals an axon branch-specific fine-tuning mechanism that locally coordinates axon remodeling and myelination.


Subject(s)
Axons , Motor Neurons/metabolism , Myelin Sheath/metabolism , Animals , Mice , Mice, Transgenic , Synaptic Transmission
6.
Front Cell Neurosci ; 12: 195, 2018.
Article in English | MEDLINE | ID: mdl-30065631

ABSTRACT

Migration and pathfinding of neuronal growth cones during neurite extension is critically dependent on dynamic microtubules. In this study we sought to determine, which aspects of microtubule polymerization relate to growth cone morphology and migratory characteristics. We conducted a multiscale quantitative microscopy analysis using automated tracking of microtubule plus ends in migrating growth cones of cultured murine dorsal root ganglion (DRG) neurons. Notably, this comprehensive analysis failed to identify any changes in microtubule polymerization parameters that were specifically associated with spontaneous extension vs. retraction of growth cones. This suggests that microtubule dynamicity is a basic mechanism that does not determine the polarity of growth cone response but can be exploited to accommodate diverse growth cone behaviors. At the same time, we found a correlation between growth cone size and basic parameters of microtubule polymerization including the density of growing microtubule plus ends and rate and duration of microtubule growth. A similar correlation was observed in growth cones of neurons lacking the microtubule-associated protein MAP1B. However, MAP1B-null growth cones, which are deficient in growth cone migration and steering, displayed an overall reduction in microtubule dynamicity. Our results highlight the importance of taking growth cone size into account when evaluating the influence on growth cone microtubule dynamics of different substrata, guidance factors or genetic manipulations which all can change growth cone morphology and size. The type of large scale multiparametric analysis performed here can help to separate direct effects that these perturbations might have on microtubule dynamics from indirect effects resulting from perturbation-induced changes in growth cone size.

7.
Neuron ; 92(4): 845-856, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27773584

ABSTRACT

Developmental axon remodeling is characterized by the selective removal of branches from axon arbors. The mechanisms that underlie such branch loss are largely unknown. Additionally, how neuronal resources are specifically assigned to the branches of remodeling arbors is not understood. Here we show that axon branch loss at the developing mouse neuromuscular junction is mediated by branch-specific microtubule severing, which results in local disassembly of the microtubule cytoskeleton and loss of axonal transport in branches that will subsequently dismantle. Accordingly, pharmacological microtubule stabilization delays neuromuscular synapse elimination. This branch-specific disassembly of the cytoskeleton appears to be mediated by the microtubule-severing enzyme spastin, which is dysfunctional in some forms of upper motor neuron disease. Our results demonstrate a physiological role for a neurodegeneration-associated modulator of the cytoskeleton, reveal unexpected cell biology of branch-specific axon plasticity and underscore the mechanistic similarities of axon loss in development and disease.


Subject(s)
Adenosine Triphosphatases/metabolism , Axonal Transport , Microtubules/metabolism , Neuromuscular Junction/metabolism , Neuronal Plasticity , Adenosine Triphosphatases/genetics , Animals , Cytoskeleton/metabolism , Mice , Mice, Knockout , Motor Neuron Disease/metabolism , Spastin
8.
Ann Neurol ; 79(5): 794-805, 2016 May.
Article in English | MEDLINE | ID: mdl-26946517

ABSTRACT

OBJECTIVE: Neuromyelitis optica (NMO) is an autoimmune disease of the central nervous system, which resembles multiple sclerosis (MS). NMO differs from MS, however, in the distribution and histology of neuroinflammatory lesions and shows a more aggressive clinical course. Moreover, the majority of NMO patients carry immunoglobulin G autoantibodies against aquaporin-4 (AQP4), an astrocytic water channel. Antibodies against AQP4 can damage astrocytes by complement, but NMO histopathology also shows demyelination, and - importantly-axon injury, which may determine permanent deficits following NMO relapses. The dynamics of astrocyte injury in NMO and the mechanisms by which toxicity spreads to axons are not understood. METHODS: Here, we establish in vivo imaging of the spinal cord, one of the main sites of NMO pathology, as a powerful tool to study the formation of experimental NMO-related lesions caused by human AQP4 antibodies in mice. RESULTS: We found that human AQP4 antibodies caused acute astrocyte depletion with initial oligodendrocyte survival. Within 2 hours of antibody application, we observed secondary axon injury in the form of progressive swellings. Astrocyte toxicity and axon damage were dependent on AQP4 antibody titer and complement, specifically C1q. INTERPRETATION: In vivo imaging of the spinal cord reveals the swift development of NMO-related acute axon injury after AQP4 antibody-mediated astrocyte depletion. This approach will be useful in studying the mechanisms underlying the spread of NMO pathology beyond astrocytes, as well as in evaluating potential neuroprotective interventions. Ann Neurol 2016;79:794-805.

9.
Arthropod Struct Dev ; 45(1): 23-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26597904

ABSTRACT

In the early embryonic grasshopper, two pairs of sibling cells near the apex of the antenna pioneer its dorsal and ventral nerve tracts to the brain. En route, the growth cones of these pioneers contact a so-called base pioneer associated with each tract and which acts as a guidepost cell. Both apical and basal pioneers express stereotypic molecular labels allowing them to be uniquely identified. Although their developmental origins are largely understood, the fates of the respective pioneers remain unclear. We therefore employed the established cell death markers acridine orange and TUNEL to determine whether the apical and basal pioneers undergo apoptosis during embryogenesis. Our data reveal that the apical pioneers maintain a consistent molecular profile from their birth up to mid-embryogenesis, at which point the initial antennal nerve tracts to the brain have been established. Shortly after this the apical pioneers undergo apoptosis. Death occurs at a developmental stage similar to that reported elsewhere for pioneers in a leg - an homologous appendage. Base pioneers, by contrast, progressively change their molecular profile and can no longer be unequivocally identified after mid-embryogenesis. At no stage up to then do they exhibit death labels. If they persist, the base pioneers must be assumed to adopt a new role in the developing antennal nervous system.


Subject(s)
Grasshoppers/embryology , Acridine Orange , Animals , Apoptosis , Arthropod Antennae/embryology , Arthropod Antennae/ultrastructure , Embryo, Nonmammalian/ultrastructure , Embryonic Development , Grasshoppers/ultrastructure , In Situ Nick-End Labeling , Neurons/ultrastructure
10.
Dev Genes Evol ; 225(3): 187-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25868908

ABSTRACT

The antennae of the grasshopper Schistocerca gregaria possess a pair of nerve pathways which are established by so-called pioneer neurons early in embryonic development. Subsequently, sensory cell clusters mediating olfaction, flight, optomotor responses, and phase changes differentiate from the antennal epithelium at stereotypic locations and direct their axons onto those of the pioneers to then project to the brain. Early in embryonic development, before the antennae become cuticularized, immunolabeling can be used to follow axogenesis in these pioneers and sensory cells. At later stages, immunolabeling becomes problematical as the cuticle is laid down and masks internal antigen sites. In order to immunolabel the nervous system of cuticularized late embryonic and first instar grasshopper antennae, we modified a procedure known as sonication in which the appendage is exposed to ultrasound thereby rendering it porous to antibodies. Comparisons of the immunolabeled nervous system of sectioned and sonicated antennae show that the cellular organization of sensory clusters and their axon projections is intact. The expression patterns of neuron-specific, microtubule-specific, and proliferative cell-specific labels in treated antennae are consistent with those reported for earlier developmental stages where sonication is not necessary, suggesting that these molecular epitopes are also conserved. The method ensures reliable immunolabeling in intact, cuticularized appendages so that the peripheral nervous system can be reconstructed directly via confocal microscopy throughout development.


Subject(s)
Grasshoppers/physiology , Immunohistochemistry , Neurons/physiology , Animals , Arthropod Antennae , Axons , Extremities , Nervous System , Neurons/cytology
11.
Neuron ; 84(6): 1183-90, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25433639

ABSTRACT

Impaired axonal transport can contribute to axon degeneration and has been described in many neurodegenerative diseases. Multiple sclerosis (MS) is a common neuroinflammatory disease, which is characterized by progressive axon degeneration-whether, when, and how axonal transport is affected in this condition is unknown. Here we used in vivo two-photon imaging to directly assay transport of organelles and the stability of microtubule tracks in individual spinal axons in mouse models of MS. We found widespread transport deficits, which preceded structural alterations of axons, cargos, or microtubules and could be reversed by acute anti-inflammatory interventions or redox scavenging. Our study shows that acute neuroinflammation induces a pervasive state of reversible axonal dysfunction, which coincides with acute disease symptoms. Moreover, perpetuated transport dysfunction, as we found in a model of progressive MS, led to reduced distal organelle supply and could thus contribute to axonal dystrophy in advanced stages of the disease.


Subject(s)
Axonal Transport/physiology , Axons/physiology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Animals , Axonal Transport/drug effects , Disease Models, Animal , Free Radical Scavengers/pharmacology , Functional Neuroimaging , Mice , Microtubules/physiology , Nerve Degeneration/physiopathology , Nitric Oxide Donors/pharmacology , Organelles/physiology , Spermine/analogs & derivatives , Spermine/pharmacology , Spinal Cord/physiology
12.
Nat Commun ; 5: 4827, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25219969

ABSTRACT

Microtubule dynamics in neurons play critical roles in physiology, injury and disease and determine microtubule orientation, the cell biological correlate of neurite polarization. Several microtubule binding proteins, including end-binding protein 3 (EB3), specifically bind to the growing plus tip of microtubules. In the past, fluorescently tagged end-binding proteins have revealed microtubule dynamics in vitro and in non-mammalian model organisms. Here, we devise an imaging assay based on transgenic mice expressing yellow fluorescent protein-tagged EB3 to study microtubules in intact mammalian neurites. Our approach allows measurement of microtubule dynamics in vivo and ex vivo in peripheral nervous system and central nervous system neurites under physiological conditions and after exposure to microtubule-modifying drugs. We find an increase in dynamic microtubules after injury and in neurodegenerative disease states, before axons show morphological indications of degeneration or regrowth. Thus increased microtubule dynamics might serve as a general indicator of neurite remodelling in health and disease.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Biological Assay , Microtubules/ultrastructure , Molecular Imaging/methods , Neurons/ultrastructure , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Polarity , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Expression , Hippocampus/cytology , Hippocampus/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neurons/metabolism , Primary Cell Culture , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Video Recording
13.
Commun Integr Biol ; 4(4): 486-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21966579

ABSTRACT

Despite a long history of anatomical mapping of neuronal networks, we are only beginning to understand the detailed three-dimensional (3D) organization of the cortical micro-circuitry. This is in part due to the lack of complete reconstructions of individual cortical neurons. Morphological studies are either performed on incomplete cells in vitro, or when performed in vivo, lack the necessary cellular resolution. We recently reconstructed the in vivo axonal and dendritic morphology of two types of L(ayer) 5 neurons from vibrissal cortex. The 3D profiles of short-range as well as longrange projections indicate that L5 slender-tufted and L5 thick-tufted neurons represent very different building blocks of the cortical circuitry. In this addendum to Oberlaender et al. (PNAS 2011), we motivate our technical approach and the advancements this may give in reconstructing the cortical micro-circuitry.

14.
Proc Natl Acad Sci U S A ; 108(10): 4188-93, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21368112

ABSTRACT

The cortical output layer 5 contains two excitatory cell types, slender- and thick-tufted neurons. In rat vibrissal cortex, slender-tufted neurons carry motion and phase information during active whisking, but remain inactive after passive whisker touch. In contrast, thick-tufted neurons reliably increase spiking preferably after passive touch. By reconstructing the 3D patterns of intracortical axon projections from individual slender- and thick-tufted neurons, filled in vivo with biocytin, we were able to identify cell type-specific intracortical circuits that may encode whisker motion and touch. Individual slender-tufted neurons showed elaborate and dense innervation of supragranular layers of large portions of the vibrissal area (total length, 86.8 ± 5.5 mm). During active whisking, these long-range projections may modulate and phase-lock the membrane potential of dendrites in layers 2 and 3 to the whisking cycle. Thick-tufted neurons with soma locations intermingling with those of slender-tufted ones display less dense intracortical axon projections (total length, 31.6 ± 14.3 mm) that are primarily confined to infragranular layers. Based on anatomical reconstructions and previous measurements of spiking, we put forward the hypothesis that thick-tufted neurons in rat vibrissal cortex receive input of whisker motion from slender-tufted neurons onto their apical tuft dendrites and input of whisker touch from thalamic neurons onto their basal dendrites. During tactile-driven behavior, such as object location, near-coincident input from these two pathways may result in increased spiking activity of thick-tufted neurons and thus enhanced signaling to their subcortical targets.


Subject(s)
Axons , Cerebral Cortex/physiology , Neurons/cytology , Vibrissae/physiology , Action Potentials , Animals , Cerebral Cortex/cytology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...