Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Adv ; 7(44): eabh0050, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34705495

ABSTRACT

The nervous system is shielded from circulating immune cells by the blood-brain barrier (BBB). During infections and autoimmune diseases, macrophages can enter the brain where they participate in pathogen elimination but can also cause tissue damage. Here, we establish a Drosophila model to study macrophage invasion into the inflamed brain. We show that the immune deficiency (Imd) pathway, but not the Toll pathway, is responsible for attraction and invasion of hemolymph-borne macrophages across the BBB during pupal stages. Macrophage recruitment is mediated by glial, but not neuronal, induction of the Imd pathway through expression of Pvf2. Within the brain, macrophages can phagocytose synaptic material and reduce locomotor abilities and longevity. Similarly, we show that central nervous system infection by group B Streptococcus elicits macrophage recruitment in an Imd-dependent manner. This suggests that evolutionarily conserved inflammatory responses require a delicate balance between beneficial and detrimental activities.

2.
J Neurosci ; 41(5): 823-833, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33468571

ABSTRACT

Phagocytic activity of glial cells is essential for proper nervous system sculpting, maintenance of circuitry, and long-term brain health. Glial engulfment of apoptotic cells and superfluous connections ensures that neuronal connections are appropriately refined, while clearance of damaged projections and neurotoxic proteins in the mature brain protects against inflammatory insults. Comparative work across species and cell types in recent years highlights the striking conservation of pathways that govern glial engulfment. Many signaling cascades used during developmental pruning are re-employed in the mature brain to "fine tune" synaptic architecture and even clear neuronal debris following traumatic events. Moreover, the neuron-glia signaling events required to trigger and perform phagocytic responses are impressively conserved between invertebrates and vertebrates. This review offers a compare-and-contrast portrayal of recent findings that underscore the value of investigating glial engulfment mechanisms in a wide range of species and contexts.


Subject(s)
Brain/cytology , Brain/growth & development , Cell Communication/physiology , Neuroglia/physiology , Neurons/physiology , Phagocytosis/physiology , Animals , Humans , Species Specificity
3.
Methods Mol Biol ; 2143: 321-338, 2020.
Article in English | MEDLINE | ID: mdl-32524491

ABSTRACT

Axon degeneration elicits a range of immune responses from local glial cells, including striking changes in glial gene expression, morphology, and phagocytic activity. Here, we describe a detailed set of protocols to assess discrete components of the glial reaction to axotomy in the adult nervous system of Drosophila melanogaster. These methods allow one to visualize and quantify transcriptional, morphological, and functional responses of glia to degenerating axons in a model system that is highly amenable to genetic manipulation.


Subject(s)
Axons/physiology , Drosophila melanogaster/immunology , Intravital Microscopy , Microscopy, Confocal/methods , Neuroglia/immunology , Wallerian Degeneration/immunology , Animals , Axotomy , Central Nervous System/pathology , DNA, Complementary/genetics , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Expression Regulation , Genes, Reporter , Image Processing, Computer-Assisted , Immunohistochemistry/instrumentation , Immunohistochemistry/methods , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroglia/metabolism , Phagocytosis , Polymerase Chain Reaction/methods , Wallerian Degeneration/physiopathology
4.
Dev Biol ; 437(1): 27-40, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29518376

ABSTRACT

Neural basic helix-loop helix (bHLH) transcription factors promote progenitor cell differentiation by activation of downstream target genes that coordinate neuronal differentiation. Here we characterize a neural bHLH target gene in Xenopus laevis, vexin (vxn; previously sbt1), that is homologous to human c8orf46 and is conserved across vertebrate species. C8orf46 has been implicated in cancer progression, but its function is unknown. Vxn is transiently expressed in differentiating progenitors in the developing central nervous system (CNS), and is required for neurogenesis in the neural plate and retina. Its function is conserved, since overexpression of either Xenopus or mouse vxn expands primary neurogenesis and promotes early retinal cell differentiation in cooperation with neural bHLH factors. Vxn protein is localized to the cell membrane and the nucleus, but functions in the nucleus to promote neural differentiation. Vxn inhibits cell proliferation, and works with the cyclin-dependent kinase inhibitor p27Xic1 (cdkn1b) to enhance neurogenesis and increase levels of the proneural protein Neurog2. We propose that vxn provides a key link between neural bHLH activity and execution of the neurogenic program.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Neurogenesis/genetics , Xenopus Proteins/genetics , Animals , Blotting, Western , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Nerve Tissue Proteins/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Retina/embryology , Retina/metabolism , Xenopus laevis
5.
J Neurosci ; 37(49): 11881-11893, 2017 12 06.
Article in English | MEDLINE | ID: mdl-29109235

ABSTRACT

Pathological hallmarks of Alzheimer's disease (AD) include amyloid-ß (Aß) plaques, neurofibrillary tangles, and reactive gliosis. Glial cells offer protection against AD by engulfing extracellular Aß peptides, but the repertoire of molecules required for glial recognition and destruction of Aß are still unclear. Here, we show that the highly conserved glial engulfment receptor Draper/MEGF10 provides neuroprotection in an AD model of Drosophila (both sexes). Neuronal expression of human Aß42arc in adult flies results in robust Aß accumulation, neurodegeneration, locomotor dysfunction, and reduced lifespan. Notably, all of these phenotypes are more severe in draper mutant animals, whereas enhanced expression of glial Draper reverses Aß accumulation, as well as behavioral phenotypes. We also show that the signal transducer and activator of transcription (Stat92E), c-Jun N-terminal kinase (JNK)/AP-1 signaling, and expression of matrix metalloproteinase-1 (Mmp1) are activated downstream of Draper in glia in response to Aß42arc exposure. Furthermore, Aß42-induced upregulation of the phagolysosomal markers Atg8 and p62 was notably reduced in draper mutant flies. Based on our findings, we propose that glia clear neurotoxic Aß peptides in the AD model Drosophila brain through a Draper/STAT92E/JNK cascade that may be coupled to protein degradation pathways such as autophagy or more traditional phagolysosomal destruction methods.SIGNIFICANCE STATEMENT Alzheimer's disease (AD) and similar dementias are common incurable neurodegenerative disorders in the aging population. As the primary immune responders in the brain, glial cells are implicated as key players in the onset and progression of AD and related disorders. Here we show that the glial engulfment receptor Draper is protective in a Drosophila model of AD, reducing levels of amyloid ß (Aß) peptides, reversing locomotor defects, and extending lifespan. We further show that protein degradation pathways are induced downstream of Draper in AD model flies, supporting a model in which glia engulf and destroy Aß peptides to reduce amyloid-associated toxicity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Neuroglia/metabolism , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Alzheimer Disease/pathology , Animals , Animals, Genetically Modified , Drosophila , Female , Male , Mice , Neuroglia/drug effects , Neuroglia/pathology
6.
Curr Opin Neurobiol ; 47: 162-167, 2017 12.
Article in English | MEDLINE | ID: mdl-29096245

ABSTRACT

Glial cells are essential for proper formation and maintenance of the nervous system. During development, glia keep neuronal cell numbers in check and ensure that mature neural circuits are appropriately sculpted by engulfing superfluous cells and projections. In the adult brain, glial cells offer metabolic sustenance and provide critical immune support in the face of acute and chronic challenges. Dysfunctional glial immune activity is believed to contribute to age-related cognitive decline, as well as neurodegenerative disease risk, but we still know surprisingly little about the specific molecular pathways that govern glia-neuron communication in the healthy or diseased brain. Drosophila offers a versatile in vivo model to explore the conserved molecular underpinnings of glial cell biology and glial cell contributions to brain function, health, and disease susceptibility. This review addresses recent findings describing how Drosophila glial cells influence neuronal activity in the adult fly brain to support optimal brain function and, importantly, highlights new insights into specific glial defects that may contribute to neuronal demise.


Subject(s)
Brain/physiology , Immunity, Innate/physiology , Neuroglia/physiology , Neurons/physiology , Animals , Drosophila
7.
Elife ; 62017 08 21.
Article in English | MEDLINE | ID: mdl-28825401

ABSTRACT

Neural injury triggers swift responses from glia, including glial migration and phagocytic clearance of damaged neurons. The transcriptional programs governing these complex innate glial immune responses are still unclear. Here, we describe a novel injury assay in adult Drosophila that elicits widespread glial responses in the ventral nerve cord (VNC). We profiled injury-induced changes in VNC gene expression by RNA sequencing (RNA-seq) and found that responsive genes fall into diverse signaling classes. One factor, matrix metalloproteinase-1 (MMP-1), is induced in Drosophila ensheathing glia responding to severed axons. Interestingly, glial induction of MMP-1 requires the highly conserved engulfment receptor Draper, as well as AP-1 and STAT92E. In MMP-1 depleted flies, glia do not properly infiltrate neuropil regions after axotomy and, as a consequence, fail to clear degenerating axonal debris. This work identifies Draper-dependent activation of MMP-1 as a novel cascade required for proper glial clearance of severed axons.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/physiology , Matrix Metalloproteinase 1/metabolism , Membrane Proteins/metabolism , Neuroglia/physiology , Peripheral Nerve Injuries/physiopathology , Signal Transduction , Animals , Disease Models, Animal , Gene Expression Profiling , STAT Transcription Factors/metabolism , Sequence Analysis, RNA , Transcription Factor AP-1/metabolism
8.
J Cell Biol ; 216(3): 531-533, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28228533

ABSTRACT

Defective immune system function is implicated in autism spectrum disorders, including Fragile X syndrome. In this issue, O'Connor et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201607093) demonstrate that phagocytic activity of systemic immune cells is compromised in a Drosophila melanogaster model of Fragile X, highlighting intriguing new mechanistic connections between FMRP, innate immunity, and abnormal development.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/immunology , Fragile X Syndrome/metabolism , Immunity, Innate/immunology , Phagocytes/immunology , Phagocytes/metabolism , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/metabolism
9.
Cell Death Dis ; 8(2): e2623, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28230857

ABSTRACT

Neuronal damage induced by injury, stroke, or neurodegenerative disease elicits swift immune responses from glial cells, including altered gene expression, directed migration to injury sites, and glial clearance of damaged neurons through phagocytic engulfment. Collectively, these responses hinder further cellular damage, but the mechanisms that underlie these important protective glial reactions are still unclear. Here, we show that the evolutionarily conserved trimeric protein phosphatase 4 (PP4) serine/threonine phosphatase complex is a novel set of factors required for proper glial responses to nerve injury in the adult Drosophila brain. Glial-specific knockdown of PP4 results in reduced recruitment of glia to severed axons and delayed glial clearance of degenerating axonal debris. We show that PP4 functions downstream of the the glial engulfment receptor Draper to drive glial morphogenesis through the guanine nucleotide exchange factor SOS and the Rho GTPase Rac1, revealing that PP4 molecularly couples Draper to Rac1-mediated cytoskeletal remodeling to ensure glial infiltration of injury sites and timely removal of damaged neurons from the CNS.


Subject(s)
Axons/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Neuroglia/metabolism , Phagocytes/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Brain/metabolism , Gene Expression Regulation/physiology , Membrane Proteins/metabolism , Neurons/metabolism , Phagocytosis/physiology , Signal Transduction/physiology
10.
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
11.
Nat Commun ; 7: 12871, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27647497

ABSTRACT

Advanced age is the greatest risk factor for neurodegenerative disorders, but the mechanisms that render the senescent brain vulnerable to disease are unclear. Glial immune responses provide neuroprotection in a variety of contexts. Thus, we explored how glial responses to neurodegeneration are altered with age. Here we show that glia-axon phagocytic interactions change dramatically in the aged Drosophila brain. Aged glia clear degenerating axons slowly due to low phosphoinositide-3-kinase (PI3K) signalling and, subsequently, reduced expression of the conserved phagocytic receptor Draper/MEGF10. Importantly, boosting PI3K/Draper activity in aged glia significantly reverses slow phagocytic responses. Moreover, several hours post axotomy, early hallmarks of Wallerian degeneration (WD) are delayed in aged flies. We propose that slow clearance of degenerating axons is mechanistically twofold, resulting from deferred initiation of axonal WD and reduced PI3K/Draper-dependent glial phagocytic function. Interventions that boost glial engulfment activity, however, can substantially reverse delayed clearance of damaged neuronal debris.


Subject(s)
Aging/physiology , Axons/metabolism , Drosophila Proteins/metabolism , Drosophila/physiology , Membrane Proteins/metabolism , Neuroglia/physiology , Animals , Axotomy , Drosophila Proteins/genetics , Membrane Proteins/genetics , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , STAT Transcription Factors/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Cell Rep ; 16(7): 1838-50, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27498858

ABSTRACT

Neuronal injury triggers robust responses from glial cells, including altered gene expression and enhanced phagocytic activity to ensure prompt removal of damaged neurons. The molecular underpinnings of glial responses to trauma remain unclear. Here, we find that the evolutionarily conserved insulin-like signaling (ILS) pathway promotes glial phagocytic clearance of degenerating axons in adult Drosophila. We find that the insulin-like receptor (InR) and downstream effector Akt1 are acutely activated in local ensheathing glia after axotomy and are required for proper clearance of axonal debris. InR/Akt1 activity, it is also essential for injury-induced activation of STAT92E and its transcriptional target draper, which encodes a conserved receptor essential for glial engulfment of degenerating axons. Increasing Draper levels in adult glia partially rescues delayed clearance of severed axons in glial InR-inhibited flies. We propose that ILS functions as a key post-injury communication relay to activate glial responses, including phagocytic activity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Insulin/metabolism , Membrane Proteins/genetics , Neuroglia/metabolism , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Animals , Axotomy , Cell Communication , Drosophila Proteins/deficiency , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Neuroglia/cytology , Neurons/pathology , Olfactory Nerve/surgery , Phagocytosis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/deficiency , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
13.
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
14.
Nat Neurosci ; 15(5): 722-30, 2012 Mar 18.
Article in English | MEDLINE | ID: mdl-22426252

ABSTRACT

Neuronal injury elicits potent cellular responses from glia, but molecular pathways modulating glial activation, phagocytic function and termination of reactive responses remain poorly defined. Here we show that positive or negative regulation of glial responses to axon injury is molecularly encoded by unique isoforms of the Drosophila melanogaster engulfment receptor Draper. Draper-I promotes engulfment of axonal debris through an immunoreceptor tyrosine-based activation motif (ITAM). In contrast, Draper-II, an alternative splice variant, potently inhibits glial engulfment function. Draper-II suppresses Draper-I signaling through a previously undescribed immunoreceptor tyrosine-based inhibitory motif (ITIM)-like domain and the tyrosine phosphatase Corkscrew (Csw). Intriguingly, loss of Draper-II-Csw signaling prolongs expression of glial engulfment genes after axotomy and reduces the ability of glia to respond to secondary axotomy. Our work highlights a novel role for Draper-II in inhibiting glial responses to neurodegeneration, and indicates that a balance of opposing Draper-I and Draper-II signaling events is essential to maintain glial sensitivity to brain injury.


Subject(s)
Axons/physiology , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Nerve Degeneration/metabolism , Neuroglia/physiology , Neurons/cytology , Animals , Animals, Genetically Modified , Apoptosis/genetics , Apoptosis/physiology , Axotomy , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Membrane Proteins/genetics , Neuroglia/cytology , Olfactory Pathways/cytology , Olfactory Pathways/injuries , Phagocytosis/genetics , Phagocytosis/physiology , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , RNA Interference/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/metabolism
15.
Nature ; 465(7301): 1093-6, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20577216

ABSTRACT

Autophagy degrades cytoplasmic components that are required for cell survival in response to starvation. Autophagy has also been associated with cell death, but it is unclear how this is distinguished from autophagy during cell survival. Drosophila salivary glands undergo programmed cell death that requires autophagy genes, and engulfment of salivary gland cells by phagocytes does not appear to occur. Here we show that Draper (Drpr), the Drosophila melanogaster orthologue of the Caenorhabditis elegans engulfment receptor CED-1, is required for autophagy during cell death. Null mutations in, and salivary gland-specific knockdown of, drpr inhibit salivary gland degradation. Knockdown of drpr prevents the induction of autophagy in dying salivary glands, and expression of the Atg1 autophagy regulator in drpr mutants suppresses the failure in degradation of salivary glands. Surprisingly, drpr is required in the same dying salivary gland cells in which it regulates autophagy induction, but drpr knockdown does not prevent starvation-induced autophagy in the fat body, which is associated with survival. In addition, components of the conserved engulfment pathway are required for clearance of dying salivary glands. To our knowledge, this is the first example of an engulfment factor that is required for self-clearance of cells. Further, Drpr is the first factor that distinguishes autophagy that is associated with cell death from autophagy associated with cell survival.


Subject(s)
Autophagy/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Animals , Animals, Genetically Modified , Autophagy/genetics , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Caspases/metabolism , Cell Death/physiology , Cell Survival , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Fat Body/cytology , Food Deprivation , Genes, Insect/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Salivary Glands/cytology , Salivary Glands/metabolism
16.
PLoS Biol ; 7(8): e1000184, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19707574

ABSTRACT

Synapse remodeling is an extremely dynamic process, often regulated by neural activity. Here we show during activity-dependent synaptic growth at the Drosophila NMJ many immature synaptic boutons fail to form stable postsynaptic contacts, are selectively shed from the parent arbor, and degenerate or disappear from the neuromuscular junction (NMJ). Surprisingly, we also observe the widespread appearance of presynaptically derived "debris" during normal synaptic growth. The shedding of both immature boutons and presynaptic debris is enhanced by high-frequency stimulation of motorneurons, indicating that their formation is modulated by neural activity. Interestingly, we find that glia dynamically invade the NMJ and, working together with muscle cells, phagocytose shed presynaptic material. Suppressing engulfment activity in glia or muscle by disrupting the Draper/Ced-6 pathway results in a dramatic accumulation of presynaptic debris, and synaptic growth in turn is severely compromised. Thus actively growing NMJ arbors appear to constitutively generate an excessive number of immature boutons, eliminate those that are not stabilized through a shedding process, and normal synaptic expansion requires the continuous clearance of this material by both glia and muscle cells.


Subject(s)
Muscles/cytology , Neuroglia/cytology , Neuromuscular Junction/cytology , Synapses/physiology , Animals , Drosophila/cytology , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/metabolism , Larva/cytology , Larva/growth & development , Larva/metabolism , Membrane Proteins/metabolism , Microscopy, Confocal , Muscles/physiology , Neuroglia/physiology , Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Reverse Transcriptase Polymerase Chain Reaction , Synapses/metabolism , Synaptic Transmission
17.
J Neurosci ; 29(15): 4768-81, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19369546

ABSTRACT

The mammalian brain contains many subtypes of glia that vary in their morphologies, gene expression profiles, and functional roles; however, the functional diversity of glia in the adult Drosophila brain remains poorly defined. Here we define the diversity of glial subtypes that exist in the adult Drosophila brain, show they bear striking similarity to mammalian brain glia, and identify the major phagocytic cell type responsible for engulfing degenerating axons after acute axotomy. We find that neuropil regions contain two different populations of glia: ensheathing glia and astrocytes. Ensheathing glia enwrap major structures in the adult brain, but are not closely associated with synapses. Interestingly, we find these glia uniquely express key components of the glial phagocytic machinery (e.g., the engulfment receptor Draper, and dCed-6), respond morphologically to axon injury, and autonomously require components of the Draper signaling pathway for successful clearance of degenerating axons from the injured brain. Astrocytic glia, in contrast, do not express Draper or dCed-6, fail to respond morphologically to axon injury, and appear to play no role in clearance of degenerating axons from the brain. However, astrocytic glia are closely associated with synaptic regions in neuropil, and express excitatory amino acid transporters, which are presumably required for the clearance of excess neurotransmitters at the synaptic cleft. Together these results argue that ensheathing glia and astrocytes are preprogrammed cell types in the adult Drosophila brain, with ensheathing glia acting as phagocytes after axotomy, and astrocytes potentially modulating synapse formation and signaling.


Subject(s)
Brain/growth & development , Myelin Sheath/physiology , Neuroglia/physiology , Phagocytes/physiology , Age Factors , Animals , Brain/cytology , Drosophila , Neuroglia/cytology , Phagocytes/cytology
18.
Neuron Glia Biol ; 3(1): 63-74, 2007 Feb.
Article in English | MEDLINE | ID: mdl-18172512

ABSTRACT

Glial cells provide support and protection for neurons in the embryonic and adult brain, mediated in part through the phagocytic activity of glia. Glial cells engulf apoptotic cells and pruned neurites from the developing nervous system, and also clear degenerating neuronal debris from the adult brain after neural trauma. Studies indicate that Drosophila melanogaster is an ideal model system to elucidate the mechanisms of engulfment by glia. The recent studies reviewed here show that many features of glial engulfment are conserved across species and argue that work in Drosophila will provide valuable cellular and molecular insight into glial engulfment activity in mammals.

19.
Dev Dyn ; 234(3): 802-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16217738

ABSTRACT

Synaptic vesicle-associated proteins are important regulators of neurotransmitter release at synaptic terminals in mature animals. Some synaptic vesicle-associated proteins are also expressed during development, although their contribution to development is not as clear. Here, we describe the cloning and developmental expression pattern of the Xenopus laevis synaptic vesicle-associated protein SVOP, a gene first identified as an immediate target for proneural basic helix-loop-helix factors. Alignment analysis revealed a high level of identity between the SVOP protein sequences from Xenopus and other vertebrates. In developing Xenopus embryos, SVOP expression is restricted to the nervous system and is first detectable at the mid-neurula stage. As development progresses SVOP becomes broadly expressed throughout the central nervous system. Our observation that SVOP is expressed in the developing Xenopus nervous system suggests that it may be involved in neuron formation, maturation, or neuronal function.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Nerve Tissue Proteins/metabolism , Nervous System/growth & development , Nervous System/metabolism , Synaptic Vesicles/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/growth & development , Xenopus laevis/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Phylogeny , Sequence Alignment , Sequence Homology, Amino Acid , Xenopus Proteins/chemistry , Xenopus Proteins/genetics
20.
Dev Biol ; 285(2): 570-83, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16112102

ABSTRACT

Proneural basic helix-loop-helix (bHLH) transcription factors are critical positive regulators of neuronal differentiation in a variety of species and are required for proper differentiation of various subtypes of neurons. Although bHLH factors demonstrate some unique functions during neural development, they share the ability to regulate neuronal differentiation, potentially by targeting overlapping sets of genes. To assess this, we performed a screen in ectoderm animal cap tissue to identify direct transcriptional targets shared by two Xenopus ato-related bHLH factors, Xath5 and XNeuroD. Candidate target genes identified in this screen include several transcriptional regulators (Xebf2, Xebf3, XETOR and NKL), an RNA binding protein (elrC), a cell cycle component (Xgadd45gamma) and several novel genes. Overexpression of either Xath5 or XNeuroD induced ectopic in vivo expression of these candidate target genes. Conversely, blocking ato-related bHLH activity prevented endogenous nervous system expression of these genes. Therefore, we have identified a set of genes that can be regulated by multiple ato-related bHLH factors and may function as critical effectors of proneural bHLH-mediated differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Ectoderm/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Genes/genetics , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA Primers , Eye Proteins/genetics , Gene Expression Profiling , In Situ Hybridization , Nerve Tissue Proteins/genetics , Nervous System/metabolism , Polymerase Chain Reaction , Xenopus/genetics , Xenopus Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...