Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36399182

ABSTRACT

Maintaining long, energetically demanding axons throughout the life of an animal is a major challenge for the nervous system. Specialized glia ensheathe axons and support their function and integrity throughout life, but glial support mechanisms remain poorly defined. Here, we identified a collection of secreted and transmembrane molecules required in glia for long-term axon survival in vivo. We showed that the majority of components of the TGFß superfamily are required in glia for sensory neuron maintenance but not glial ensheathment of axons. In the absence of glial TGFß signaling, neurons undergo age-dependent degeneration that can be rescued either by genetic blockade of Wallerian degeneration or caspase-dependent death. Blockade of glial TGFß signaling results in increased ATP in glia that can be mimicked by enhancing glial mitochondrial biogenesis or suppressing glial monocarboxylate transporter function. We propose that glial TGFß signaling supports axon survival and suppresses neurodegeneration through promoting glial metabolic support of neurons.


Subject(s)
Axons , Neuroglia , Transforming Growth Factor beta , Animals , Axons/metabolism , Neuroglia/metabolism , Peripheral Nerves/cytology , Sensory Receptor Cells , Transforming Growth Factor beta/metabolism , Drosophila melanogaster , Organelle Biogenesis , Monocarboxylic Acid Transporters/metabolism
2.
Development ; 149(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36355066

ABSTRACT

Most invertebrate axons and small-caliber axons in mammalian peripheral nerves are unmyelinated but still ensheathed by glia. Here, we use Drosophila wrapping glia to study the development and function of non-myelinating axon ensheathment, which is poorly understood. Selective ablation of these glia from peripheral nerves severely impaired larval locomotor behavior. In an in vivo RNA interference screen to identify glial genes required for axon ensheathment, we identified the conserved receptor tyrosine kinase Discoidin domain receptor (Ddr). In larval peripheral nerves, loss of Ddr resulted in severely reduced ensheathment of axons and reduced axon caliber, and we found a strong dominant genetic interaction between Ddr and the type XV/XVIII collagen Multiplexin (Mp), suggesting that Ddr functions as a collagen receptor to drive axon wrapping. In adult nerves, loss of Ddr decreased long-term survival of sensory neurons and significantly reduced axon caliber without overtly affecting ensheathment. Our data establish essential roles for non-myelinating glia in nerve development, maintenance and function, and identify Ddr as a key regulator of axon-glia interactions during ensheathment and establishment of axon caliber.


Subject(s)
Axons , Drosophila Proteins , Animals , Discoidin Domain Receptors , Axons/physiology , Neuroglia , Drosophila Proteins/genetics , Peripheral Nerves , Drosophila , Mammals
3.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33972422

ABSTRACT

There is a tight association between mitochondrial dysfunction and neurodegenerative diseases and axons that are particularly vulnerable to degeneration, but how mitochondria are maintained in axons to support their physiology remains poorly defined. In an in vivo forward genetic screen for mutants altering axonal mitochondria, we identified tsg101 Neurons mutant for tsg101 exhibited an increase in mitochondrial number and decrease in mitochondrial size. TSG101 is best known as a component of the endosomal sorting complexes required for transport (ESCRT) complexes; however, loss of most other ESCRT components did not affect mitochondrial numbers or size, suggesting TSG101 regulates mitochondrial biology in a noncanonical, ESCRT-independent manner. The TSG101-mutant phenotype was not caused by lack of mitophagy, and we found that autophagy blockade was detrimental only to the mitochondria in the cell bodies, arguing mitophagy and autophagy are dispensable for the regulation of mitochondria number in axons. Interestingly, TSG101 mitochondrial phenotypes were instead caused by activation of PGC-1ɑ/Nrf2-dependent mitochondrial biogenesis, which was mTOR independent and TFEB dependent and required the mitochondrial fission-fusion machinery. Our work identifies a role for TSG101 in inhibiting mitochondrial biogenesis, which is essential for the maintenance of mitochondrial numbers and sizes, in the axonal compartment.


Subject(s)
Axons/metabolism , DNA-Binding Proteins/genetics , Drosophila melanogaster/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Mitochondria/genetics , Organelle Biogenesis , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Male , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitophagy/genetics , Mutation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/cytology , Neurons/metabolism , Transcription Factors/metabolism
4.
Curr Biol ; 29(23): 3961-3973.e6, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31735672

ABSTRACT

Transcripts of noxious stimulus-detecting TrpA1 channels are alternatively spliced. Despite the importance of nociception for survival, the in vivo significance of expressing different TrpA1 isoforms is largely unknown. Here, we develop a novel genetic approach to generate Drosophila knockin strains expressing single TrpA1 isoforms. Drosophila TrpA1 mediates heat and UVC-triggered nociception. We show that TrpA1-C and TrpA1-D, two alternative isoforms, are co-expressed in nociceptors. When examined in heterologous cells, both TrpA1-C and TrpA1-D are activated by heat and UVC. By contrast, analysis of knockin flies reveals the striking functional specificity; TrpA1-C mediates UVC-nociception, whereas TrpA1-D mediates heat-nociception. Therefore, in vivo functions of TrpA1-C and TrpA1-D are different from each other and are different from their in vitro properties. Our results indicate that a given sensory stimulus preferentially activates a single TrpA1 isoform in vivo and that polymodal nociception requires co-expression of TrpA1 isoforms, providing novel insights of how alternative splicing regulates nociception.


Subject(s)
Alternative Splicing , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Ion Channels/genetics , Nociception , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Ion Channels/metabolism , Protein Isoforms/genetics , Single-Cell Analysis
5.
Neuron ; 103(1): 52-65.e6, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31101394

ABSTRACT

Mitochondria are essential in long axons to provide metabolic support and sustain neuron integrity. A healthy mitochondrial pool is maintained by biogenesis, transport, mitophagy, fission, and fusion, but how these events are regulated in axons is not well defined. Here, we show that the Drosophila glutathione S-transferase (GST) Gfzf prevents mitochondrial hyperfusion in axons. Gfzf loss altered redox balance between glutathione (GSH) and oxidized glutathione (GSSG) and initiated mitochondrial fusion through the coordinated action of Mfn and Opa1. Gfzf functioned epistatically with the thioredoxin peroxidase Jafrac1 and the thioredoxin reductase 1 TrxR-1 to regulate mitochondrial dynamics. Altering GSH:GSSG ratios in mouse primary neurons in vitro also induced hyperfusion. Mitochondrial changes caused deficits in trafficking, the metabolome, and neuronal physiology. Changes in GSH and oxidative state are associated with neurodegenerative diseases like Alzheimer's. Our demonstration that GSTs are key in vivo regulators of axonal mitochondrial length and number provides a potential mechanistic link.


Subject(s)
Axons/physiology , Carrier Proteins/physiology , Glutathione/metabolism , Mitochondria/physiology , Animals , Axons/ultrastructure , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Female , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Neurons/metabolism , Oxidation-Reduction , Peroxidases/genetics , Peroxidases/physiology , Pregnancy , Primary Cell Culture , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/physiology
6.
Genes Dev ; 31(20): 2023-2038, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29138279

ABSTRACT

Most glial functions depend on establishing intimate morphological relationships with neurons. Significant progress has been made in understanding neuron-glia signaling at synaptic and axonal contacts, but how glia support neuronal cell bodies is unclear. Here we explored the growth and functions of Drosophila cortex glia (which associate almost exclusively with neuronal cell bodies) to understand glia-soma interactions. We show that cortex glia tile with one another and with astrocytes to establish unique central nervous system (CNS) spatial domains that actively restrict glial growth, and selective ablation of cortex glia causes animal lethality. In an RNAi-based screen, we identified αSNAP (soluble NSF [N-ethylmalemeide-sensitive factor] attachment protein α) and several components of vesicle fusion and recycling machinery as essential for the maintenance of cortex glial morphology and continued contact with neurons. Interestingly, loss of the secreted neurotrophin Spätzle 3 (Spz3) phenocopied αSNAP phenotypes, which included loss of glial ensheathment of neuron cell bodies, increased neuronal cell death, and defects in animal behavior. Rescue experiments suggest that Spz3 can exert these effects only over very short distances. This work identifies essential roles for glial ensheathment of neuronal cell bodies in CNS homeostasis as well as Spz3 as a novel signaling factor required for maintenance of cortex glial morphology and neuron-glia contact.


Subject(s)
Brain/embryology , Drosophila Proteins/physiology , Neuroglia/cytology , Neurons/cytology , Animals , Astrocytes/cytology , Behavior, Animal , Brain/cytology , Brain/growth & development , Cell Survival , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Membrane Fusion , Morphogenesis , RNA Interference
7.
Neuron ; 95(1): 78-91.e5, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683272

ABSTRACT

Axon degeneration is a hallmark of neurodegenerative disease and neural injury. Axotomy activates an intrinsic pro-degenerative axon death signaling cascade involving loss of the NAD+ biosynthetic enzyme Nmnat/Nmnat2 in axons, activation of dSarm/Sarm1, and subsequent Sarm-dependent depletion of NAD+. Here we identify Axundead (Axed) as a mediator of axon death. axed mutants suppress axon death in several types of axons for the lifespan of the fly and block the pro-degenerative effects of activated dSarm in vivo. Neurodegeneration induced by loss of the sole fly Nmnat ortholog is also fully blocked by axed, but not dsarm, mutants. Thus, pro-degenerative pathways activated by dSarm signaling or Nmnat elimination ultimately converge on Axed. Remarkably, severed axons morphologically preserved by axon death pathway mutations remain integrated in circuits and able to elicit complex behaviors after stimulation, indicating that blockade of axon death signaling results in long-term functional preservation of axons.


Subject(s)
Armadillo Domain Proteins/genetics , Axons/metabolism , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Wallerian Degeneration/genetics , Animals , Animals, Genetically Modified , Armadillo Domain Proteins/metabolism , Arthropod Antennae/injuries , Arthropod Antennae/innervation , Axotomy , Behavior, Animal , Blotting, Western , Cell Line , Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Grooming , Immunity, Active , NAD/metabolism , Neurons/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Optogenetics , Wallerian Degeneration/metabolism , Wings, Animal/injuries , Wings, Animal/innervation
8.
Nat Commun ; 8: 14355, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165006

ABSTRACT

Draper/Ced-1/MEGF-10 is an engulfment receptor that promotes clearance of cellular debris in C. elegans, Drosophila and mammals. Draper signals through an evolutionarily conserved Src family kinase cascade to drive cytoskeletal rearrangements and target engulfment through Rac1. Glia also alter gene expression patterns in response to axonal injury but pathways mediating these responses are poorly defined. We show Draper is cell autonomously required for glial activation of transcriptional reporters after axonal injury. We identify TNF receptor associated factor 4 (TRAF4) as a novel Draper binding partner that is required for reporter activation and phagocytosis of axonal debris. TRAF4 and misshapen (MSN) act downstream of Draper to activate c-Jun N-terminal kinase (JNK) signalling in glia, resulting in changes in transcriptional reporters that are dependent on Drosophila AP-1 (dAP-1) and STAT92E. Our data argue injury signals received by Draper at the membrane are important regulators of downstream transcriptional responses in reactive glia.


Subject(s)
Axons/pathology , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Nerve Degeneration/metabolism , Neuroglia/pathology , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Axons/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Drosophila melanogaster/metabolism , Female , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Nerve Degeneration/pathology , Neuroglia/cytology , Neuroglia/metabolism , Phagocytosis , STAT Transcription Factors/metabolism , TNF Receptor-Associated Factor 4/metabolism , Transcription Factor AP-1/metabolism
9.
PLoS Biol ; 12(11): e1001985, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25369313

ABSTRACT

Glial cells are exquisitely sensitive to neuronal injury but mechanisms by which glia establish competence to respond to injury, continuously gauge neuronal health, and rapidly activate reactive responses remain poorly defined. Here, we show glial PI3K signaling in the uninjured brain regulates baseline levels of Draper, a receptor essential for Drosophila glia to sense and respond to axonal injury. After injury, Draper levels are up-regulated through a Stat92E-modulated, injury-responsive enhancer element within the draper gene. Surprisingly, canonical JAK/STAT signaling does not regulate draper expression. Rather, we find injury-induced draper activation is downstream of the Draper/Src42a/Shark/Rac1 engulfment signaling pathway. Thus, PI3K signaling and Stat92E are critical in vivo regulators of glial responsiveness to axonal injury. We provide evidence for a positive auto-regulatory mechanism whereby signaling through the injury-responsive Draper receptor leads to Stat92E-dependent, transcriptional activation of the draper gene. We propose that Drosophila glia use this auto-regulatory loop as a mechanism to adjust their reactive state following injury.


Subject(s)
Diffuse Axonal Injury/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Neuroglia/physiology , Phosphatidylinositol 3-Kinases/metabolism , STAT Transcription Factors/metabolism , Animals , Axons/metabolism , Brain/metabolism , Drosophila , Drosophila Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Janus Kinases/metabolism , Membrane Proteins/genetics , Olfactory Receptor Neurons/physiology , Signal Transduction
10.
Science ; 337(6093): 481-4, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22678360

ABSTRACT

Axonal and synaptic degeneration is a hallmark of peripheral neuropathy, brain injury, and neurodegenerative disease. Axonal degeneration has been proposed to be mediated by an active autodestruction program, akin to apoptotic cell death; however, loss-of-function mutations capable of potently blocking axon self-destruction have not been described. Here, we show that loss of the Drosophila Toll receptor adaptor dSarm (sterile α/Armadillo/Toll-Interleukin receptor homology domain protein) cell-autonomously suppresses Wallerian degeneration for weeks after axotomy. Severed mouse Sarm1 null axons exhibit remarkable long-term survival both in vivo and in vitro, indicating that Sarm1 prodegenerative signaling is conserved in mammals. Our results provide direct evidence that axons actively promote their own destruction after injury and identify dSarm/Sarm1 as a member of an ancient axon death signaling pathway.


Subject(s)
Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/physiology , Axons/physiology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Neurons/physiology , Wallerian Degeneration , Animals , Animals, Genetically Modified , Apoptosis , Armadillo Domain Proteins/analysis , Axons/ultrastructure , Axotomy , Cell Survival , Cells, Cultured , Cytoskeletal Proteins/analysis , Denervation , Drosophila/embryology , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/analysis , Mice , Mutation , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Signal Transduction , Superior Cervical Ganglion/cytology , Tissue Culture Techniques
11.
J Cell Biol ; 184(4): 501-13, 2009 Feb 23.
Article in English | MEDLINE | ID: mdl-19237597

ABSTRACT

Slow Wallerian degeneration (Wld(S)) encodes a chimeric Ube4b/nicotinamide mononucleotide adenylyl transferase 1 (Nmnat1) fusion protein that potently suppresses Wallerian degeneration, but the mechanistic action of Wld(S) remains controversial. In this study, we characterize Wld(S)-mediated axon protection in vivo using Drosophila melanogaster. We show that Nmnat1 can protect severed axons from autodestruction but at levels significantly lower than Wld(S), and enzyme-dead versions of Nmnat1 and Wld(S) exhibit severely reduced axon-protective function. Interestingly, a 16-amino acid N-terminal domain of Wld(S) (termed N16) accounts for the differences in axon-sparing activity between Wld(S) and Nmnat1, and N16-dependent enhancement of Nmnat1-protective activity in Wld(S) requires the N16-binding protein valosin-containing protein (VCP)/TER94. Thus, Wld(S)-mediated suppression of Wallerian degeneration results from VCP-N16 interactions and Nmnat1 activity converging in vivo. Surprisingly, mouse Nmnat3, a mitochondrial Nmnat enzyme that localizes to the cytoplasm in Drosophila cells, protects severed axons at levels indistinguishable from Wld(S). Thus, nuclear Nmnat activity does not appear to be essential for Wld(S)-like axon protection.


Subject(s)
Axons/metabolism , Nerve Tissue Proteins/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals , Animals, Genetically Modified , Drosophila melanogaster , Mice
12.
Nature ; 453(7197): 935-9, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18432193

ABSTRACT

The cellular machinery promoting phagocytosis of corpses of apoptotic cells is well conserved from worms to mammals. An important component is the Caenorhabditis elegans engulfment receptor CED-1 (ref. 1) and its Drosophila orthologue, Draper. The CED-1/Draper signalling pathway is also essential for the phagocytosis of other types of 'modified self' including necrotic cells, developmentally pruned axons and dendrites, and axons undergoing Wallerian degeneration. Here we show that Drosophila Shark, a non-receptor tyrosine kinase similar to mammalian Syk and Zap-70, binds Draper through an immunoreceptor tyrosine-based activation motif (ITAM) in the Draper intracellular domain. We show that Shark activity is essential for Draper-mediated signalling events in vivo, including the recruitment of glial membranes to severed axons and the phagocytosis of axonal debris and neuronal cell corpses by glia. We also show that the Src family kinase (SFK) Src42A can markedly increase Draper phosphorylation and is essential for glial phagocytic activity. We propose that ligand-dependent Draper receptor activation initiates the Src42A-dependent tyrosine phosphorylation of Draper, the association of Shark and the activation of the Draper pathway. These Draper-Src42A-Shark interactions are strikingly similar to mammalian immunoreceptor-SFK-Syk signalling events in mammalian myeloid and lymphoid cells. Thus, Draper seems to be an ancient immunoreceptor with an extracellular domain tuned to modified self, and an intracellular domain promoting phagocytosis through an ITAM-domain-SFK-Syk-mediated signalling cascade.


Subject(s)
Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neuroglia/cytology , Phagocytosis , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Axons/metabolism , Axons/pathology , Cell Line , Cell Membrane/metabolism , Central Nervous System , Drosophila Proteins/chemistry , Membrane Proteins/chemistry , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein Transport , Syk Kinase , Two-Hybrid System Techniques
13.
Neuron ; 50(6): 869-81, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16772169

ABSTRACT

Neuron-glia communication is central to all nervous system responses to trauma, yet neural injury signaling pathways remain poorly understood. Here we explore cellular and molecular aspects of neural injury signaling in Drosophila. We show that transected Drosophila axons undergo injury-induced degeneration that is morphologically similar to Wallerian degeneration in mammals and can be suppressed by the neuroprotective mouse Wlds protein. Axonal injury elicits potent morphological and molecular responses from Drosophila glia: glia upregulate expression of the engulfment receptor Draper, undergo dramatic changes in morphology, and rapidly recruit cellular processes toward severed axons. In draper mutants, glia fail to respond morphologically to axon injury, and severed axons are not cleared from the CNS. Thus Draper appears to act as a glial receptor for severed axon-derived molecular cues that drive recruitment of glial processes to injured axons for engulfment.


Subject(s)
Axons/physiology , Drosophila Proteins/physiology , Membrane Proteins/physiology , Neuroglia/physiology , Animals , Animals, Genetically Modified , Drosophila , Nerve Tissue Proteins/physiology , Neuroglia/cytology , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Wallerian Degeneration/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...