1
|
Lee JY, Gala DS, Kiourlappou M, Olivares-Abril J, Joha J, Titlow JS, Teodoro RO, Davis I. Murine glial protrusion transcripts predict localized Drosophila glial mRNAs involved in plasticity. J Cell Biol 2024; 223:e202306152. [PMID: 39037431 PMCID: PMC11262410 DOI: 10.1083/jcb.202306152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
The polarization of cells often involves the transport of specific mRNAs and their localized translation in distal projections. Neurons and glia are both known to contain long cytoplasmic processes, while localized transcripts have only been studied extensively in neurons, not glia, especially in intact nervous systems. Here, we predict 1,740 localized Drosophila glial transcripts by extrapolating from our meta-analysis of seven existing studies characterizing the localized transcriptomes and translatomes of synaptically associated mammalian glia. We demonstrate that the localization of mRNAs in mammalian glial projections strongly predicts the localization of their high-confidence Drosophila homologs in larval motor neuron-associated glial projections and are highly statistically enriched for genes associated with neurological diseases. We further show that some of these localized glial transcripts are specifically required in glia for structural plasticity at the nearby neuromuscular junction synapses. We conclude that peripheral glial mRNA localization is a common and conserved phenomenon and propose that it is likely to be functionally important in disease.
Collapse
Affiliation(s)
- Jeffrey Y. Lee
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Dalia S. Gala
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | | | - Jana Joha
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Rita O. Teodoro
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ilan Davis
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Clayworth K, Gilbert M, Auld V. Cell Biology Techniques for Studying Drosophila Peripheral Glial Cells. Cold Spring Harb Protoc 2024; 2024:pdb.top108159. [PMID: 37399179 DOI: 10.1101/pdb.top108159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
Glial cells are essential for the proper development and functioning of the peripheral nervous system (PNS). The ability to study the biology of glial cells is therefore critical for our ability to understand PNS biology and address PNS maladies. The genetic and proteomic pathways underlying vertebrate peripheral glial biology are understandably complex, with many layers of redundancy making it sometimes difficult to study certain facets of PNS biology. Fortunately, many aspects of vertebrate peripheral glial biology are conserved with those of the fruit fly, Drosophila melanogaster With simple and powerful genetic tools and fast generation times, Drosophila presents an accessible and versatile model for studying the biology of peripheral glia. We introduce here three techniques for studying the cell biology of peripheral glia of Drosophila third-instar larvae. With fine dissection tools and common laboratory reagents, third-instar larvae can be dissected, with extraneous tissues removed, revealing the central nervous system (CNS) and PNS to be processed using a standard immunolabeling protocol. To improve the resolution of peripheral nerves in the z-plane, we describe a cryosectioning method to achieve 10- to 20-µm thick coronal sections of whole larvae, which can then be immunolabeled using a modified version of standard immunolabeling techniques. Finally, we describe a proximity ligation assay (PLA) for detecting close proximity between two proteins-thus inferring protein interaction-in vivo in third-instar larvae. These methods, further described in our associated protocols, can be used to improve our understanding of Drosophila peripheral glia biology, and thus our understanding of PNS biology.
Collapse
Affiliation(s)
- Katherine Clayworth
- Department of Zoology, Cell and Developmental Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Mary Gilbert
- Department of Zoology, Cell and Developmental Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Vanessa Auld
- Department of Zoology, Cell and Developmental Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
3
|
Fernandes VM, Auld V, Klämbt C. Glia as Functional Barriers and Signaling Intermediaries. Cold Spring Harb Perspect Biol 2024; 16:a041423. [PMID: 38167424 PMCID: PMC10759988 DOI: 10.1101/cshperspect.a041423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Glia play a crucial role in providing metabolic support to neurons across different species. To do so, glial cells isolate distinct neuronal compartments from systemic signals and selectively transport specific metabolites and ions to support neuronal development and facilitate neuronal function. Because of their function as barriers, glial cells occupy privileged positions within the nervous system and have also evolved to serve as signaling intermediaries in various contexts. The fruit fly, Drosophila melanogaster, has significantly contributed to our understanding of glial barrier development and function. In this review, we will explore the formation of the glial sheath, blood-brain barrier, and nerve barrier, as well as the significance of glia-extracellular matrix interactions in barrier formation. Additionally, we will delve into the role of glia as signaling intermediaries in regulating nervous system development, function, and response to injury.
Collapse
Affiliation(s)
- Vilaiwan M Fernandes
- Department of Cell and Developmental Biology, University College London, London UC1E 6DE, United Kingdom
| | - Vanessa Auld
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Christian Klämbt
- Institute for Neuro- and Behavioral Biology, University of Münster, Münster 48149, Germany
| |
Collapse
|
4
|
Baldenius M, Kautzmann S, Nanda S, Klämbt C. Signaling Pathways Controlling Axonal Wrapping in Drosophila. Cells 2023; 12:2553. [PMID: 37947631 PMCID: PMC10647682 DOI: 10.3390/cells12212553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
The rapid transmission of action potentials is an important ability that enables efficient communication within the nervous system. Glial cells influence conduction velocity along axons by regulating the radial axonal diameter, providing electrical insulation as well as affecting the distribution of voltage-gated ion channels. Differentiation of these wrapping glial cells requires a complex set of neuron-glia interactions involving three basic mechanistic features. The glia must recognize the axon, grow around it, and eventually arrest its growth to form single or multiple axon wraps. This likely depends on the integration of numerous evolutionary conserved signaling and adhesion systems. Here, we summarize the mechanisms and underlying signaling pathways that control glial wrapping in Drosophila and compare those to the mechanisms that control glial differentiation in mammals. This analysis shows that Drosophila is a beneficial model to study the development of even complex structures like myelin.
Collapse
Affiliation(s)
| | | | | | - Christian Klämbt
- Institute for Neuro- and Behavioral Biology, Faculty of Biology, University of Münster, Röntgenstraße 16, D-48149 Münster, Germany; (M.B.)
| |
Collapse
|
5
|
Hopkins BR, Barmina O, Kopp A. A single-cell atlas of the sexually dimorphic Drosophila foreleg and its sensory organs during development. PLoS Biol 2023; 21:e3002148. [PMID: 37379332 DOI: 10.1371/journal.pbio.3002148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/03/2023] [Indexed: 06/30/2023] Open
Abstract
To respond to the world around them, animals rely on the input of a network of sensory organs distributed throughout the body. Distinct classes of sensory organs are specialized for the detection of specific stimuli such as strain, pressure, or taste. The features that underlie this specialization relate both to the neurons that innervate sensory organs and the accessory cells they comprise. To understand the genetic basis of this diversity of cell types, both within and between sensory organs, we performed single-cell RNA sequencing on the first tarsal segment of the male Drosophila melanogaster foreleg during pupal development. This tissue displays a wide variety of functionally and structurally distinct sensory organs, including campaniform sensilla, mechanosensory bristles, and chemosensory taste bristles, as well as the sex comb, a recently evolved male-specific structure. In this study, we characterize the cellular landscape in which the sensory organs reside, identify a novel cell type that contributes to the construction of the neural lamella, and resolve the transcriptomic differences among support cells within and between sensory organs. We identify the genes that distinguish between mechanosensory and chemosensory neurons, resolve a combinatorial transcription factor code that defines 4 distinct classes of gustatory neurons and several types of mechanosensory neurons, and match the expression of sensory receptor genes to specific neuron classes. Collectively, our work identifies core genetic features of a variety of sensory organs and provides a rich, annotated resource for studying their development and function.
Collapse
Affiliation(s)
- Ben R Hopkins
- Department of Evolution and Ecology, University of California, Davis, California, United States of America
| | - Olga Barmina
- Department of Evolution and Ecology, University of California, Davis, California, United States of America
| | - Artyom Kopp
- Department of Evolution and Ecology, University of California, Davis, California, United States of America
| |
Collapse
|
6
|
Corty MM, Coutinho-Budd J. Drosophila glia take shape to sculpt the nervous system. Curr Opin Neurobiol 2023; 79:102689. [PMID: 36822142 PMCID: PMC10023329 DOI: 10.1016/j.conb.2023.102689] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 02/23/2023]
Abstract
The importance of glial cells has become increasingly apparent over the past 20 years, yet compared to neurons we still know relatively little about these essential cells. Most critical glial cell functions are conserved in Drosophila glia, often using the same key molecular players as their vertebrate counterparts. The relative simplicity of the Drosophila nervous system, combined with a vast array of powerful genetic tools, allows us to further dissect the molecular composition and functional roles of glia in ways that would be much more cumbersome or not possible in higher vertebrate systems. Importantly, Drosophila genetics allow for in vivo manipulation, and their transparent body wall enables in vivo imaging of glia in intact animals throughout early development. Here we discuss recent advances in Drosophila glial development detailing how these cells take on their mature morphologies and interact with neurons to perform their important functional roles in the nervous system.
Collapse
Affiliation(s)
- Megan M Corty
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA. https://twitter.com/@megancphd
| | - Jaeda Coutinho-Budd
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
7
|
Das M, Cheng D, Matzat T, Auld VJ. Innexin-Mediated Adhesion between Glia Is Required for Axon Ensheathment in the Peripheral Nervous System. J Neurosci 2023; 43:2260-2276. [PMID: 36801823 PMCID: PMC10072304 DOI: 10.1523/jneurosci.1323-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 01/03/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Glia are essential to protecting and enabling nervous system function and a key glial function is the formation of the glial sheath around peripheral axons. Each peripheral nerve in the Drosophila larva is ensheathed by three glial layers, which structurally support and insulate the peripheral axons. How peripheral glia communicate with each other and between layers is not well established and we investigated the role of Innexins in mediating glial function in the Drosophila periphery. Of the eight Drosophila Innexins, we found two (Inx1 and Inx2) are important for peripheral glia development. In particular loss of Inx1 and Inx2 resulted in defects in the wrapping glia leading to disruption of the glia wrap. Of interest loss of Inx2 in the subperineurial glia also resulted in defects in the neighboring wrapping glia. Inx plaques were observed between the subperineurial glia and the wrapping glia suggesting that gap junctions link these two glial cell types. We found Inx2 is key to Ca2+ pulses in the peripheral subperineurial glia but not in the wrapping glia, and we found no evidence of gap junction communication between subperineurial and wrapping glia. Rather we have clear evidence that Inx2 plays an adhesive and channel-independent role between the subperineurial and wrapping glia to ensure the integrity of the glial wrap.SIGNIFICANCE STATEMENT Gap junctions are critical for glia communication and formation of myelin in myelinating glia. However, the role of gap junctions in non-myelinating glia is not well studied, yet non-myelinating glia are critical for peripheral nerve function. We found the Innexin gap junction proteins are present between different classes of peripheral glia in Drosophila. Here Innexins form junctions to facilitate adhesion between the different glia but do so in a channel-independent manner. Loss of adhesion leads to disruption of the glial wrap around axons and leads to fragmentation of the wrapping glia membranes. Our work points to an important role for gap junction proteins in mediating insulation by non-myelinating glia.
Collapse
Affiliation(s)
- Mriga Das
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Duo Cheng
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Till Matzat
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Vanessa J Auld
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
8
|
Contreras EG, Klämbt C. The Drosophila blood-brain barrier emerges as a model for understanding human brain diseases. Neurobiol Dis 2023; 180:106071. [PMID: 36898613 DOI: 10.1016/j.nbd.2023.106071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.
Collapse
Affiliation(s)
- Esteban G Contreras
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| | - Christian Klämbt
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| |
Collapse
|
9
|
Corty MM, Hulegaard AL, Hill JQ, Sheehan AE, Aicher SA, Freeman MR. Discoidin domain receptor regulates ensheathment, survival and caliber of peripheral axons. Development 2022; 149:281293. [PMID: 36355066 PMCID: PMC10112903 DOI: 10.1242/dev.200636] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/27/2022] [Indexed: 11/12/2022]
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.
Collapse
Affiliation(s)
- Megan M Corty
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Jo Q Hill
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy E Sheehan
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sue A Aicher
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Marc R Freeman
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
10
|
Delta/Notch signaling in glia maintains motor nerve barrier function and synaptic transmission by controlling matrix metalloproteinase expression. Proc Natl Acad Sci U S A 2022; 119:e2110097119. [PMID: 35969789 PMCID: PMC9407389 DOI: 10.1073/pnas.2110097119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have made a surprising discovery linking Delta/Notch signaling in subperineurial glia (SPG) to the regulation of nerve ensheathment and neurotransmitter release at the Drosophila neuromuscular junction (NMJ). SPG, the counterpart of the endothelial layer in the vertebrate blood–brain barrier, form the key cellular layer that is critical for axonal ensheathment and the blood–brain barrier in Drosophila. Our findings demonstrate that Delta/Notch signaling exerts a constitutive negative inhibition on JNK signaling in SPG, thereby limiting the expression of Mmp1, a matrix metalloproteinase. SPG-specific and temporally regulated knockdown of Delta leads to breakdown of barrier function and compromises neurotransmitter release at the NMJ. Our results provide a mechanistic insight into the biology of barrier function and glia–neuron interactions. While the role of barrier function in establishing a protective, nutrient-rich, and ionically balanced environment for neurons has been appreciated for some time, little is known about how signaling cues originating in barrier-forming cells participate in maintaining barrier function and influence synaptic activity. We have identified Delta/Notch signaling in subperineurial glia (SPG), a crucial glial type for Drosophila motor axon ensheathment and the blood–brain barrier, to be essential for controlling the expression of matrix metalloproteinase 1 (Mmp1), a major regulator of the extracellular matrix (ECM). Our genetic analysis indicates that Delta/Notch signaling in SPG exerts an inhibitory control on Mmp1 expression. In the absence of this inhibition, abnormally enhanced Mmp1 activity disrupts septate junctions and glial ensheathment of peripheral motor nerves, compromising neurotransmitter release at the neuromuscular junction (NMJ). Temporally controlled and cell type–specific transgenic analysis shows that Delta/Notch signaling inhibits transcription of Mmp1 by inhibiting c-Jun N-terminal kinase (JNK) signaling in SPG. Our results provide a mechanistic insight into the regulation of neuronal health and function via glial-initiated signaling and open a framework for understanding the complex relationship between ECM regulation and the maintenance of barrier function.
Collapse
|
11
|
Contreras EG, Sierralta J. The Fly Blood-Brain Barrier Fights Against Nutritional Stress. Neurosci Insights 2022; 17:26331055221120252. [PMID: 36225749 PMCID: PMC9549514 DOI: 10.1177/26331055221120252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
In the wild, animals face different challenges including multiple events of food
scarcity. How they overcome these conditions is essential for survival. Thus,
adaptation mechanisms evolved to allow the development and survival of an
organism during nutrient restriction periods. Given the high energy demand of
the nervous system, the molecular mechanisms of adaptation to malnutrition are
of great relevance to fuel the brain. The blood-brain barrier (BBB) is the
interface between the central nervous system (CNS) and the circulatory system.
The BBB mediates the transport of macromolecules in and out of the CNS, and
therefore, it can buffer changes in nutrient availability. In this review, we
collect the current evidence using the fruit fly, Drosophila
melanogaster, as a model of the role of the BBB in the adaptation
to starvation. We discuss the role of the Drosophila BBB during
nutrient deprivation as a potential sensor for circulating nutrients, and
transient nutrient storage as a regulator of the CNS neurogenic niche.
Collapse
Affiliation(s)
- Esteban G Contreras
- Institute of Neuro- and Behavioral Biology, University of Münster, Münster, Germany
| | - Jimena Sierralta
- Biomedical Neuroscience Institute and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
12
|
Weiss S, Clamon LC, Manoim JE, Ormerod KG, Parnas M, Littleton JT. Glial ER and GAP junction mediated Ca 2+ waves are crucial to maintain normal brain excitability. Glia 2021; 70:123-144. [PMID: 34528727 DOI: 10.1002/glia.24092] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 11/08/2022]
Abstract
Astrocytes play key roles in regulating multiple aspects of neuronal function from invertebrates to humans and display Ca2+ fluctuations that are heterogeneously distributed throughout different cellular microdomains. Changes in Ca2+ dynamics represent a key mechanism for how astrocytes modulate neuronal activity. An unresolved issue is the origin and contribution of specific glial Ca2+ signaling components at distinct astrocytic domains to neuronal physiology and brain function. The Drosophila model system offers a simple nervous system that is highly amenable to cell-specific genetic manipulations to characterize the role of glial Ca2+ signaling. Here we identify a role for ER store-operated Ca2+ entry (SOCE) pathway in perineurial glia (PG), a glial population that contributes to the Drosophila blood-brain barrier. We show that PG cells display diverse Ca2+ activity that varies based on their locale within the brain. Ca2+ signaling in PG cells does not require extracellular Ca2+ and is blocked by inhibition of SOCE, Ryanodine receptors, or gap junctions. Disruption of these components triggers stimuli-induced seizure-like episodes. These findings indicate that Ca2+ release from internal stores and its propagation between neighboring glial cells via gap junctions are essential for maintaining normal nervous system function.
Collapse
Affiliation(s)
- Shirley Weiss
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lauren C Clamon
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Julia E Manoim
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kiel G Ormerod
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Moshe Parnas
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
Krill JL, Dawson-Scully K. Characterization of a novel stimulus-induced glial calcium wave in Drosophila larval peripheral segmental nerves and its role in PKG-modulated thermoprotection. J Neurogenet 2021; 35:221-235. [PMID: 34309496 DOI: 10.1080/01677063.2021.1941945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Insects, as poikilotherms, have adaptations to deal with wide ranges in temperature fluctuation. Allelic variations in the foraging gene that encodes a cGMP dependent protein kinase, were discovered to have effects on behavior in Drosophila by Dr. Marla Sokolowski in 1980. This single gene has many pleiotropic effects and influences feeding behavior, metabolic storage, learning and memory and has been shown to affect stress tolerance. PKG regulation affects motoneuronal thermotolerance in Drosophila larvae as well as adults. While the focus of thermotolerance studies has been on the modulation of neuronal function, other cell types have been overlooked. Because glia are vital to neuronal function and survival, we wanted to determine if glia play a role in thermotolerance as well. In our investigation, we discovered a novel calcium wave at the larval NMJ and set out to characterize the wave's dynamics and the potential mechanism underlying the wave prior to determining what effect, if any, PKG modulation has on the thermotolerance of glia cells. Using pharmacology, we determined that calcium buffering mechanisms of the mitochondria and endoplasmic reticulum play a role in the propagation of our novel glial calcium wave. By coupling pharmacology with genetic manipulation using RNA interference (RNAi), we found that PKG modulation in glia alters thermoprotection of function as well as glial calcium wave dynamics.
Collapse
Affiliation(s)
- Jennifer L Krill
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida
| | - Ken Dawson-Scully
- Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida
| |
Collapse
|
14
|
Hertenstein H, McMullen E, Weiler A, Volkenhoff A, Becker HM, Schirmeier S. Starvation-induced regulation of carbohydrate transport at the blood-brain barrier is TGF-β-signaling dependent. eLife 2021; 10:e62503. [PMID: 34032568 PMCID: PMC8149124 DOI: 10.7554/elife.62503] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
During hunger or malnutrition, animals prioritize alimentation of the brain over other organs to ensure its function and, thus, their survival. This protection, also-called brain sparing, is described from Drosophila to humans. However, little is known about the molecular mechanisms adapting carbohydrate transport. Here, we used Drosophila genetics to unravel the mechanisms operating at the blood-brain barrier (BBB) under nutrient restriction. During starvation, expression of the carbohydrate transporter Tret1-1 is increased to provide more efficient carbohydrate uptake. Two mechanisms are responsible for this increase. Similar to the regulation of mammalian GLUT4, Rab-dependent intracellular shuttling is needed for Tret1-1 integration into the plasma membrane; even though Tret1-1 regulation is independent of insulin signaling. In addition, starvation induces transcriptional upregulation that is controlled by TGF-β signaling. Considering TGF-β-dependent regulation of the glucose transporter GLUT1 in murine chondrocytes, our study reveals an evolutionarily conserved regulatory paradigm adapting the expression of sugar transporters at the BBB.
Collapse
Affiliation(s)
- Helen Hertenstein
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Ellen McMullen
- Institut für Neuro- und Verhaltensbiologie, WWU MünsterMünsterGermany
| | - Astrid Weiler
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Anne Volkenhoff
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Holger M Becker
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
- Division of General Zoology, Department of Biology, University of KaiserslauternKaiserslauternGermany
| | - Stefanie Schirmeier
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| |
Collapse
|
15
|
Wrapping glia regulates neuronal signaling speed and precision in the peripheral nervous system of Drosophila. Nat Commun 2020; 11:4491. [PMID: 32901033 PMCID: PMC7479103 DOI: 10.1038/s41467-020-18291-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
The functionality of the nervous system requires transmission of information along axons with high speed and precision. Conductance velocity depends on axonal diameter whereas signaling precision requires a block of electrical crosstalk between axons, known as ephaptic coupling. Here, we use the peripheral nervous system of Drosophila larvae to determine how glia regulates axonal properties. We show that wrapping glial differentiation depends on gap junctions and FGF-signaling. Abnormal glial differentiation affects axonal diameter and conductance velocity and causes mild behavioral phenotypes that can be rescued by a sphingosine-rich diet. Ablation of wrapping glia does not further impair axonal diameter and conductance velocity but causes a prominent locomotion phenotype that cannot be rescued by sphingosine. Moreover, optogenetically evoked locomotor patterns do not depend on conductance speed but require the presence of wrapping glial processes. In conclusion, our data indicate that wrapping glia modulates both speed and precision of neuronal signaling.
Collapse
|
16
|
Basigin Associates with Integrin in Order to Regulate Perineurial Glia and Drosophila Nervous System Morphology. J Neurosci 2020; 40:3360-3373. [PMID: 32265259 DOI: 10.1523/jneurosci.1397-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
The Drosophila nervous system is ensheathed by a layer of outer glial cells, the perineurial glia, and a specialized extracellular matrix, the neural lamella. The function of perineurial glial cells and how they interact with the extracellular matrix are just beginning to be elucidated. Integrin-based focal adhesion complexes link the glial membrane to the extracellular matrix, but little is known about integrin's regulators in the glia. The transmembrane Ig domain protein Basigin/CD147/EMMPRIN is highly expressed in the perineurial glia surrounding the Drosophila larval nervous system. Here we show that Basigin associates with integrin at the focal adhesions to uphold the structure of the glia-extracellular matrix sheath. Knockdown of Basigin in perineurial glia using RNAi results in significant shortening of the ventral nerve cord, compression of the glia and extracellular matrix in the peripheral nerves, and reduction in larval locomotion. We determined that Basigin is expressed in close proximity to integrin at the glial membrane, and that expression of the extracellular integrin-binding domain of Basigin is sufficient to rescue peripheral glial compression. We also found that a reduction in expression of integrin at the membrane rescues the ventral nerve cord shortening, peripheral glial compression, and locomotor phenotypes, and that reduction in the integrin-binding protein Talin can partially rescue glial compression. These results identify Basigin as a potential negative regulator of integrin in the glia, supporting proper glial and extracellular matrix ensheathment of the nervous system.SIGNIFICANCE STATEMENT The glial cells and extracellular matrix play important roles in supporting and protecting the nervous system, but the interactions between these components have not been well characterized. Our study identified expression of a conserved Ig superfamily protein, Basigin, at the glial membrane of Drosophila where it associates with the integrin-based focal adhesion complexes to ensure proper ensheathment of the CNS and PNS. Loss of Basigin in the glia results in an overall compression of the nervous system due to integrin dysregulation, which causes locomotor defects in the animals. This underlies the importance of glia-matrix communication for structural and functional support of the nervous system.
Collapse
|
17
|
Silva-Rodrigues JF, Patrício-Rodrigues CF, de Sousa-Xavier V, Augusto PM, Fernandes AC, Farinho AR, Martins JP, Teodoro RO. Peripheral axonal ensheathment is regulated by RalA GTPase and the exocyst complex. Development 2020; 147:dev.174540. [PMID: 31969325 DOI: 10.1242/dev.174540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/14/2020] [Indexed: 12/21/2022]
Abstract
Axon ensheathment is fundamental for fast impulse conduction and the normal physiological functioning of the nervous system. Defects in axonal insulation lead to debilitating conditions, but, despite its importance, the molecular players responsible are poorly defined. Here, we identify RalA GTPase as a key player in axon ensheathment in Drosophila larval peripheral nerves. We demonstrate through genetic analysis that RalA action through the exocyst complex is required in wrapping glial cells to regulate their growth and development. We suggest that the RalA-exocyst pathway controls the targeting of secretory vesicles for membrane growth or for the secretion of a wrapping glia-derived factor that itself regulates growth. In summary, our findings provide a new molecular understanding of the process by which axons are ensheathed in vivo, a process that is crucial for normal neuronal function.
Collapse
Affiliation(s)
- Joana F Silva-Rodrigues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Cátia F Patrício-Rodrigues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Vicente de Sousa-Xavier
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Pedro M Augusto
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Ana C Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Ana R Farinho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - João P Martins
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rita O Teodoro
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| |
Collapse
|
18
|
Scarpelli EM, Trinh VY, Tashnim Z, Krans JL, Keller LC, Colodner KJ. Developmental expression of human tau in Drosophila melanogaster glial cells induces motor deficits and disrupts maintenance of PNS axonal integrity, without affecting synapse formation. PLoS One 2019; 14:e0226380. [PMID: 31821364 PMCID: PMC6903755 DOI: 10.1371/journal.pone.0226380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/25/2019] [Indexed: 11/26/2022] Open
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the abnormal phosphorylation and accumulation of the microtubule-associated protein, tau, in both neuronal and glial cells. Though tau pathology in glial cells is a prominent feature of many of these disorders, the pathological contribution of these lesions to tauopathy pathogenesis remains largely unknown. Moreover, while tau pathology is predominantly found in the central nervous system, a role for tau in the cells of the peripheral nervous system has been described, though not well characterized. To investigate the effects of glial tau expression on the development and maintenance of the peripheral nervous system, we utilized a Drosophila melanogaster model of tauopathy that expresses human wild-type tau in glial cells during development. We found that glial tau expression during development results in larval locomotor deficits and organismal lethality at the pupal stage, without affecting larval neuromuscular junction synapse development or post-synaptic amplitude. There was, however, a significant decrease in the decay time of synaptic potentials upon repeated stimulation of the motoneuron. Behavioral abnormalities were accompanied by glial cell death, disrupted maintenance of glial-axonal integrity, and the abnormal accumulation of the presynaptic protein, Bruchpilot, in peripheral nerve axons. Together, these data demonstrate that human tau expression in Drosophila glial cells does not affect neuromuscular junction synapse formation during development, but is deleterious to the maintenance of glial-axonal interactions in the peripheral nervous system.
Collapse
Affiliation(s)
- Enrico M. Scarpelli
- Frank H. Netter, M.D. School of Medicine, Quinnipiac University, North Haven, CT, United States of America
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, United States of America
| | - Van Y. Trinh
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, United States of America
| | - Zarrin Tashnim
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, United States of America
| | - Jacob L. Krans
- Department of Neuroscience, Western New England University, Springfield, MA, United States of America
| | - Lani C. Keller
- Frank H. Netter, M.D. School of Medicine, Quinnipiac University, North Haven, CT, United States of America
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, United States of America
| | - Kenneth J. Colodner
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, MA, United States of America
| |
Collapse
|
19
|
Li H, Russo A, DiAntonio A. SIK3 suppresses neuronal hyperexcitability by regulating the glial capacity to buffer K + and water. J Cell Biol 2019; 218:4017-4029. [PMID: 31645458 PMCID: PMC6891094 DOI: 10.1083/jcb.201907138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/27/2019] [Accepted: 09/19/2019] [Indexed: 01/10/2023] Open
Abstract
Glial regulation of extracellular potassium (K+) helps to maintain appropriate levels of neuronal excitability. While channels and transporters mediating K+ and water transport are known, little is understood about upstream regulatory mechanisms controlling the glial capacity to buffer K+ and osmotically obliged water. Here we identify salt-inducible kinase 3 (SIK3) as the central node in a signal transduction pathway controlling glial K+ and water homeostasis in Drosophila Loss of SIK3 leads to dramatic extracellular fluid accumulation in nerves, neuronal hyperexcitability, and seizures. SIK3-dependent phenotypes are exacerbated by K+ stress. SIK3 promotes the cytosolic localization of HDAC4, thereby relieving inhibition of Mef2-dependent transcription of K+ and water transport molecules. This transcriptional program controls the glial capacity to regulate K+ and water homeostasis and modulate neuronal excitability. We identify HDAC4 as a candidate therapeutic target in this pathway, whose inhibition can enhance the K+ buffering capacity of glia, which may be useful in diseases of dysregulated K+ homeostasis and hyperexcitability.
Collapse
Affiliation(s)
- Hailun Li
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO
| | - Alexandra Russo
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University in St. Louis School of Medicine, St. Louis, MO
| |
Collapse
|
20
|
Ray M, Acharya S, Shambhavi S, Lakhotia SC. Over-expression of Hsp83 in grossly depleted hsrω lncRNA background causes synthetic lethality and l(2)gl phenocopy in Drosophila. J Biosci 2019; 44:36. [PMID: 31180049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We examined interactions between the 83 kDa heat-shock protein (Hsp83) and hsrω long noncoding RNAs (lncRNAs) in hsrω66 Hsp90GFP homozygotes, which almost completely lack hsrω lncRNAs but over-express Hsp83. All +/+; hsrω66 Hsp90GFP progeny died before the third instar. Rare Sp/CyO; hsrω66 Hsp90GFP reached the third instar stage but phenocopied l(2)gl mutants, becoming progressively bulbous and transparent with enlarged brain and died after prolonged larval life. Additionally, ventral ganglia too were elongated. However, hsrω66 Hsp90GFP/TM6B heterozygotes, carrying +/+ or Sp/CyO second chromosomes, developed normally. Total RNA sequencing (+/+, +/+; hsrω66/hsrω66, Sp/CyO; hsrω66/ hsrω66, +/+; Hsp90GFP/Hsp90GFP and Sp/CyO; hsrω66 Hsp90GFP/hsrω66 Hsp90GFP late third instar larvae) revealed similar effects on many genes in hsrω66 and Hsp90GFP homozygotes. Besides additive effect on many of them, numerous additional genes were affected in Sp/CyO; hsrω66 Hsp90GFP larvae, with l(2)gl and several genes regulating the central nervous system being highly down-regulated in surviving Sp/CyO; hsrω66 Hsp90GFP larvae, but not in hsrω66 or Hsp90GFP single mutants. Hsp83 and several omega speckle-associated hnRNPs were bioinformatically found to potentially bind with these gene promoters and transcripts. Since Hsp83 and hnRNPs are also known to interact, elevated Hsp83 in an altered background of hnRNP distribution and dynamics, due to near absence of hsrω lncRNAs and omega speckles, can severely perturb regulatory circuits with unexpected consequences, including down-regulation of tumoursuppressor genes such as l(2)gl.
Collapse
Affiliation(s)
- Mukulika Ray
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | | | | | | |
Collapse
|
21
|
Sapar ML, Han C. Die in pieces: How Drosophila sheds light on neurite degeneration and clearance. J Genet Genomics 2019; 46:187-199. [PMID: 31080046 PMCID: PMC6541534 DOI: 10.1016/j.jgg.2019.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 01/08/2023]
Abstract
Dendrites and axons are delicate neuronal membrane extensions that undergo degeneration after physical injuries. In neurodegenerative diseases, they often degenerate prior to neuronal death. Understanding the mechanisms of neurite degeneration has been an intense focus of neurobiology research in the last two decades. As a result, many discoveries have been made in the molecular pathways that lead to neurite degeneration and the cell-cell interactions responsible for the subsequent clearance of neuronal debris. Drosophila melanogaster has served as a prime in vivo model system for identifying and characterizing the key molecular players in neurite degeneration, thanks to its genetic tractability and easy access to its nervous system. The knowledge learned in the fly provided targets and fuel for studies in other model systems that have further enhanced our understanding of neurodegeneration. In this review, we will introduce the experimental systems developed in Drosophila to investigate injury-induced neurite degeneration, and then discuss the biological pathways that drive degeneration. We will also cover what is known about the mechanisms of how phagocytes recognize and clear degenerating neurites, and how recent findings in this area enhance our understanding of neurodegenerative disease pathology.
Collapse
Affiliation(s)
- Maria L Sapar
- Weill Institute for Cell and Molecular Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Chun Han
- Weill Institute for Cell and Molecular Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
22
|
Over-expression of Hsp83 in grossly depleted hsrω lncRNA background causes synthetic lethality and l(2)gl phenocopy in Drosophila. J Biosci 2019. [DOI: 10.1007/s12038-019-9852-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Glial ensheathment of the somatodendritic compartment regulates sensory neuron structure and activity. Proc Natl Acad Sci U S A 2019; 116:5126-5134. [PMID: 30804200 DOI: 10.1073/pnas.1814456116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sensory neurons perceive environmental cues and are important of organismal survival. Peripheral sensory neurons interact intimately with glial cells. While the function of axonal ensheathment by glia is well studied, less is known about the functional significance of glial interaction with the somatodendritic compartment of neurons. Herein, we show that three distinct glia cell types differentially wrap around the axonal and somatodendritic surface of the polymodal dendritic arborization (da) neuron of the Drosophila peripheral nervous system for detection of thermal, mechanical, and light stimuli. We find that glial cell-specific loss of the chromatin modifier gene dATRX in the subperineurial glial layer leads to selective elimination of somatodendritic glial ensheathment, thus allowing us to investigate the function of such ensheathment. We find that somatodendritic glial ensheathment regulates the morphology of the dendritic arbor, as well as the activity of the sensory neuron, in response to sensory stimuli. Additionally, glial ensheathment of the neuronal soma influences dendritic regeneration after injury.
Collapse
|
24
|
Hans VR, Wendt TI, Patel AM, Patel MM, Perez L, Talbot DE, Jemc JC. Raw regulates glial population of the eye imaginal disc. Genesis 2018; 56:e23254. [DOI: 10.1002/dvg.23254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/25/2018] [Accepted: 09/29/2018] [Indexed: 01/25/2023]
Affiliation(s)
| | - Taylor I. Wendt
- Department of BiologyLoyola University Chicago Chicago Illinois
| | | | - Mit M. Patel
- Department of BiologyLoyola University Chicago Chicago Illinois
| | - Luselena Perez
- Department of BiologyLoyola University Chicago Chicago Illinois
| | | | | |
Collapse
|
25
|
Yildirim K, Petri J, Kottmeier R, Klämbt C. Drosophila glia: Few cell types and many conserved functions. Glia 2018; 67:5-26. [DOI: 10.1002/glia.23459] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Kerem Yildirim
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Johanna Petri
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Rita Kottmeier
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| | - Christian Klämbt
- Institute for Neuro and Behavioral Biology; University of Münster; Badestraße 9, 48149 Münster Germany
| |
Collapse
|
26
|
Zülbahar S, Sieglitz F, Kottmeier R, Altenhein B, Rumpf S, Klämbt C. Differential expression of Öbek controls ploidy in the Drosophila blood-brain barrier. Development 2018; 145:dev.164111. [PMID: 30002129 DOI: 10.1242/dev.164111] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/03/2018] [Indexed: 12/13/2022]
Abstract
During development, tissue growth is mediated by either cell proliferation or cell growth, coupled with polyploidy. Both strategies are employed by the cell types that make up the Drosophila blood-brain barrier. During larval growth, the perineurial glia proliferate, whereas the subperineurial glia expand enormously and become polyploid. Here, we show that the level of ploidy in the subperineurial glia is controlled by the N-terminal asparagine amidohydrolase homolog Öbek, and high Öbek levels are required to limit replication. In contrast, perineurial glia express moderate levels of Öbek, and increased Öbek expression blocks their proliferation. Interestingly, other dividing cells are not affected by alteration of Öbek expression. In glia, Öbek counteracts fibroblast growth factor and Hippo signaling to differentially affect cell growth and number. We propose a mechanism by which growth signals are integrated differentially in a glia-specific manner through different levels of Öbek protein to adjust cell proliferation versus endoreplication in the blood-brain barrier.
Collapse
Affiliation(s)
- Selen Zülbahar
- Institute of Neurobiology, University of Münster, Badestrasse 9, 48149 Münster, Germany
| | - Florian Sieglitz
- Institute of Neurobiology, University of Münster, Badestrasse 9, 48149 Münster, Germany
| | - Rita Kottmeier
- Institute of Neurobiology, University of Münster, Badestrasse 9, 48149 Münster, Germany
| | - Benjamin Altenhein
- Institute of Zoology, University of Cologne, Zülpicher Straße 47b, 50674 Cologne, Germany
| | - Sebastian Rumpf
- Institute of Neurobiology, University of Münster, Badestrasse 9, 48149 Münster, Germany
| | - Christian Klämbt
- Institute of Neurobiology, University of Münster, Badestrasse 9, 48149 Münster, Germany
| |
Collapse
|
27
|
Identification of raw as a regulator of glial development. PLoS One 2018; 13:e0198161. [PMID: 29813126 PMCID: PMC5973607 DOI: 10.1371/journal.pone.0198161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 05/15/2018] [Indexed: 12/18/2022] Open
Abstract
Glial cells perform numerous functions to support neuron development and function, including axon wrapping, formation of the blood brain barrier, and enhancement of synaptic transmission. We have identified a novel gene, raw, which functions in glia of the central and peripheral nervous systems in Drosophila. Reducing Raw levels in glia results in morphological defects in the brain and ventral nerve cord, as well as defects in neuron function, as revealed by decreased locomotion in crawling assays. Examination of the number of glia along peripheral nerves reveals a reduction in glial number upon raw knockdown. The reduced number of glia along peripheral nerves occurs as a result of decreased glial proliferation. As Raw has been shown to negatively regulate Jun N-terminal kinase (JNK) signaling in other developmental contexts, we examined the expression of a JNK reporter and the downstream JNK target, matrix metalloproteinase 1 (mmp1), and found that raw knockdown results in increased reporter activity and Mmp1 levels. These results are consistent with previous studies showing increased Mmp levels lead to nerve cord defects similar to those observed upon raw knockdown. In addition, knockdown of puckered, a negative feedback regulator of JNK signaling, also causes a decrease in glial number. Thus, our studies have resulted in the identification of a new regulator of gliogenesis, and demonstrate that increased JNK signaling negatively impacts glial development.
Collapse
|
28
|
Zhang K, Tan J, Su J, Liang H, Shen L, Li C, Pan G, Yang L, Cui H. Integrin β3 plays a novel role in innate immunity in silkworm, Bombyx mori. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 77:307-317. [PMID: 28826989 DOI: 10.1016/j.dci.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/16/2017] [Accepted: 08/17/2017] [Indexed: 06/07/2023]
Abstract
Integrins are transmembrane receptors that play essential roles in many physiological and pathological processes through cell-to-cell and cell-to-extracellular matrix (ECM) interactions. In the current study, a 2653-bp full-length cDNA of a novel integrin β subunit (designated Bmintegrin β3) was obtained from silkworm hemocytes. Bmintegrin β3 has the typical conserved structure of the integrin β family. The qRT-PCR results showed that Bmintegrin β3 was specifically expressed in the hematological system and that its expression was significantly increased after challenge with different types of PAMPs and bacteria. The recombinant Bmintegrin β3 protein displayed increased aggregation with S. aureus, suggesting that Bmintegrin β3 might directly bind to PAMPs. Interestingly, Bmintegrin β3 knockdown promoted PPO1, PPO2, BAEE, SPH78, SPH125, and SPH127 expression and accelerated the melanization process. Unexpectedly, the expression of genes related to phagocytosis, the Toll pathway, and the IMD pathway was also up-regulated after Bmintegrin β3 knockdown. Thus, Bmintegrin β3 might be a pattern recognition protein (PRP) for PAMPs and might directly bind to bacteria and enhance the phagocytosis activity of hemocytes. Moreover, Bmintegrin β3 and its ligand might negatively regulate the expression of immune-related genes through an unknown mechanism. In summary, our studies provide new insights into the immune functions of Bmintegrin β3 from the silkworm, Bombyx mori.
Collapse
Affiliation(s)
- Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Juan Tan
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Jingjing Su
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Hanghua Liang
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Li Shen
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Chongyang Li
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Guangzhao Pan
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing 400716, China.
| |
Collapse
|
29
|
Sasse S, Klämbt C. Repulsive Epithelial Cues Direct Glial Migration along the Nerve. Dev Cell 2017; 39:696-707. [PMID: 27997826 DOI: 10.1016/j.devcel.2016.11.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/14/2016] [Accepted: 11/19/2016] [Indexed: 11/29/2022]
Abstract
Most glial cells show pronounced migratory abilities and generally follow axonal trajectories to reach their final destination. However, the molecular cues controlling their directional migration are largely unknown. To address this, we established glial migration onto the developing Drosophila leg imaginal disc as a model. Here, CNS-derived glial cells move along nerves containing motoaxons and sensory axons. Along their path, glial cells encounter at least three choice points where directional decisions are needed. Subsequent genetic analyses allowed uncovering mechanisms that escaped previous studies. Most strikingly, we found that glial cells require the expression of the repulsive guidance receptors PlexinA/B and Robo2 to prevent breaking away from the nerve. Interestingly, the repulsive ligands are presented by the underlying leg imaginal disc epithelium, which appears to push glial cells toward the axon fascicle. In conclusion, nerve formation not only requires neuron-glia interaction but also depends on glial-epithelial communication.
Collapse
Affiliation(s)
- Sofia Sasse
- Institut für Neuro- und Verhaltensbiologie, Badestraße 9, 48149 Münster, Germany
| | - Christian Klämbt
- Institut für Neuro- und Verhaltensbiologie, Badestraße 9, 48149 Münster, Germany.
| |
Collapse
|
30
|
Schwabe T, Li X, Gaul U. Dynamic analysis of the mesenchymal-epithelial transition of blood-brain barrier forming glia in Drosophila. Biol Open 2017; 6:232-243. [PMID: 28108476 PMCID: PMC5312092 DOI: 10.1242/bio.020669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
During development, many epithelia are formed by a mesenchymal-epithelial transition (MET). Here, we examine the major stages and underlying mechanisms of MET during blood-brain barrier formation in Drosophila. We show that contact with the basal lamina is essential for the growth of the barrier-forming subperineurial glia (SPG). Septate junctions (SJs), which provide insulation of the paracellular space, are not required for MET, but are necessary for the establishment of polarized SPG membrane compartments. In vivo time-lapse imaging reveals that the Moody GPCR signaling pathway regulates SPG cell growth and shape, with different levels of signaling causing distinct phenotypes. Timely, well-coordinated SPG growth is essential for the uniform insertion of SJs and thus the insulating function of the barrier. To our knowledge, this is the first dynamic in vivo analysis of all stages in the formation of a secondary epithelium, and of the key role trimeric G protein signaling plays in this important morphogenetic process. Summary: This study examines the major steps and underlying mechanisms of mesenchymal-epithelial transition of the blood-brain-barrier forming glia in Drosophila, including the role of basal lamina, septate junctions and of trimeric G protein signaling.
Collapse
Affiliation(s)
- Tina Schwabe
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Feodor-Lynen-Str. 25, Munich 81377, Germany
| | - Xiaoling Li
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Feodor-Lynen-Str. 25, Munich 81377, Germany.,Rockefeller University, 1230 York Ave, New York, 10065-6399 NY, USA
| | - Ulrike Gaul
- Department of Biochemistry, Gene Center, Center of Integrated Protein Science (CIPSM), University of Munich, Feodor-Lynen-Str. 25, Munich 81377, Germany
| |
Collapse
|
31
|
Accumulation of Laminin Monomers in Drosophila Glia Leads to Glial Endoplasmic Reticulum Stress and Disrupted Larval Locomotion. J Neurosci 2016; 36:1151-64. [PMID: 26818504 DOI: 10.1523/jneurosci.1797-15.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The nervous system is surrounded by an extracellular matrix composed of large glycoproteins, including perlecan, collagens, and laminins. Glial cells in many organisms secrete laminin, a large heterotrimeric protein consisting of an α, β, and γ subunit. Prior studies have found that loss of laminin subunits from vertebrate Schwann cells causes loss of myelination and neuropathies, results attributed to loss of laminin-receptor signaling. We demonstrate that loss of the laminin γ subunit (LanB2) in the peripheral glia of Drosophila melanogaster results in the disruption of glial morphology due to disruption of laminin secretion. Specifically, knockdown of LanB2 in peripheral glia results in accumulation of the β subunit (LanB1), leading to distended endoplasmic reticulum (ER), ER stress, and glial swelling. The physiological consequences of disruption of laminin secretion in glia included decreased larval locomotion and ultimately lethality. Loss of the γ subunit from wrapping glia resulted in a disruption in the glial ensheathment of axons but surprisingly did not affect animal locomotion. We found that Tango1, a protein thought to exclusively mediate collagen secretion, is also important for laminin secretion in glia via a collagen-independent mechanism. However loss of secretion of the laminin trimer does not disrupt animal locomotion. Rather, it is the loss of one subunit that leads to deleterious consequences through the accumulation of the remaining subunits. SIGNIFICANCE STATEMENT This research presents a new perspective on how mutations in the extracellular matrix protein laminin cause severe consequences in glial wrapping and function. Glial-specific loss of the β or γ laminin subunit disrupted glia morphology and led to ER expansion and stress due to retention of other subunits. The retention of the unpaired laminin subunit was key to the glial disruption as loss of Tango1 blocked secretion of the complete laminin trimer but did not lead to glial or locomotion defects. The effects were observed in the perineurial glia that envelope the peripheral and central nervous systems, providing evidence for the importance of this class of glia in supporting nervous system function.
Collapse
|
32
|
Sasse S, Neuert H, Klämbt C. Differentiation ofDrosophilaglial cells. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:623-36. [DOI: 10.1002/wdev.198] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/25/2015] [Accepted: 05/24/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Sofia Sasse
- Institut für Neuro- und Verhaltensbiologie; Münster Germany
| | - Helen Neuert
- Institut für Neuro- und Verhaltensbiologie; Münster Germany
| | | |
Collapse
|
33
|
Tavares L, Pereira E, Correia A, Santos MA, Amaral N, Martins T, Relvas JB, Pereira PS. Drosophila PS2 and PS3 integrins play distinct roles in retinal photoreceptors-glia interactions. Glia 2015; 63:1155-65. [PMID: 25731761 DOI: 10.1002/glia.22806] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/28/2015] [Indexed: 11/09/2022]
Abstract
Cellular migration and differentiation are important developmental processes that require dynamic cellular adhesion. Integrins are heterodimeric transmembrane receptors that play key roles in adhesion plasticity. Here, we explore the developing visual system of Drosophila to study the roles of integrin heterodimers in glia development. Our data show that αPS2 is essential for retinal glia migration from the brain into the eye disc and that glial cells have a role in the maintenance of the fenestrated membrane (Laminin-rich ECM layer) in the disc. Interestingly, the absence of glial cells in the eye disc did not affect the targeting of retinal axons to the optic stalk. In contrast, αPS3 is not required for retinal glia migration, but together with Talin, it functions in glial cells to allow photoreceptor axons to target the optic stalk. Thus, we present evidence that αPS2 and αPS3 integrin have different and specific functions in the development of retinal glia.
Collapse
Affiliation(s)
- Lígia Tavares
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
34
|
de Luca AC, Lacour SP, Raffoul W, di Summa PG. Extracellular matrix components in peripheral nerve repair: how to affect neural cellular response and nerve regeneration? Neural Regen Res 2015; 9:1943-8. [PMID: 25598773 PMCID: PMC4283273 DOI: 10.4103/1673-5374.145366] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2014] [Indexed: 01/09/2023] Open
Abstract
Peripheral nerve injury is a serious problem affecting significantly patients’ life. Autografts are the “gold standard” used to repair the injury gap, however, only 50% of patients fully recover from the trauma. Artificial conduits are a valid alternative to repairing peripheral nerve. They aim at confining the nerve environment throughout the regeneration process, and providing guidance to axon outgrowth. Biocompatible materials have been carefully designed to reduce inflammation and scar tissue formation, but modifications of the inner lumen are still required in order to optimise the scaffolds. Biomicking the native neural tissue with extracellular matrix fillers or coatings showed great promises in repairing longer gaps and extending cell survival. In addition, extracellular matrix molecules provide a platform to further bind growth factors that can be released in the system over time. Alternatively, conduit fillers can be used for cell transplantation at the injury site, reducing the lag time required for endogenous Schwann cells to proliferate and take part in the regeneration process. This review provides an overview on the importance of extracellular matrix molecules in peripheral nerve repair.
Collapse
Affiliation(s)
- Alba C de Luca
- EPFL, Centre for Neuroprosthetics, Laboratory for Soft Bioelectronic Interfaces, Station 17, 1015 Lausanne, Switzerland
| | - Stephanie P Lacour
- EPFL, Centre for Neuroprosthetics, Laboratory for Soft Bioelectronic Interfaces, Station 17, 1015 Lausanne, Switzerland
| | - Wassim Raffoul
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Pietro G di Summa
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
35
|
Jia Q, Liu Y, Liu H, Li S. Mmp1 and Mmp2 cooperatively induce Drosophila fat body cell dissociation with distinct roles. Sci Rep 2014; 4:7535. [PMID: 25520167 PMCID: PMC4269897 DOI: 10.1038/srep07535] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 11/27/2014] [Indexed: 12/31/2022] Open
Abstract
During Drosophila metamorphosis, the single-cell layer of fat body tissues gradually dissociates into individual cells. Via a fat body-specific RNAi screen in this study, we found that two matrix metalloproteinases (MMPs), Mmp1 and Mmp2, are both required for fat body cell dissociation. As revealed through a series of cellular, biochemical, molecular, and genetic experiments, Mmp1 preferentially cleaves DE-cadherin-mediated cell-cell junctions, while Mmp2 preferentially degrades basement membrane (BM) components and thus destroy cell-BM junctions, resulting in the complete dissociation of the entire fat body tissues into individual cells. Moreover, several genetic interaction experiments demonstrated that the roles of Mmp1 and Mmp2 in this developmental process are cooperative. In conclusion, Mmp1 and Mmp2 induce fat body cell dissociation during Drosophila metamorphosis in a cooperative yet distinct manner, a finding that sheds light on the general mechanisms by which MMPs regulate tissue remodeling in animals.
Collapse
Affiliation(s)
- Qiangqiang Jia
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yang Liu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Hanhan Liu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Sheng Li
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
36
|
Katagiri N, Katagiri Y, Wada M, Okano D, Shigematsu Y, Yoshioka T. Three-dimensional reconstruction of the axon extending from the dermal photoreceptor cell in the extraocular photoreception system of a marine gastropod, onchidium. Zoolog Sci 2014; 31:810-9. [PMID: 25483793 DOI: 10.2108/zs140085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The marine gastropod Onchidium has a multiple photoreceptive system consisting of stalk eyes, dorsal eyes, photosensitive neurons, and extraocular dermal photoreceptor cells (DPCs). The DPCs were widespread all over the dorsal mantle and distributed singly or in groups in the dermis, but were not discernible by the naked eye. The DPC was oval in shape and large in size, and characterized by features specific to gastropod photoreceptor cells such as massive microvilli, photic vesicles, and a depolarized response. DPC-17, one of a group of 19 DPCs, was examined on serial semi-thin sections of 0.4 µm in thickness with a high-voltage transmission electron microscope (HVTEM). The axon emerged specifically from the lateral side between the distal microvillous portion and proximal cytoplasm, travelled through the connective tissue, and joined a small nerve bundle (NB). Two types of supportive cells were found along the length of the axon. The first type was a covering cell (CC) surrounding the surface of the DPC body and continuing onward to the axon sheath. DPC-17 was covered by 11 CCs, while the larger DPC-6 was only covered by four CCs. The second type was a sheath cell (ShC) wrapping the surface of the small NB where the axon of the DPC merged with undefined nerve fibers. The axon extending directly from DPC-17 was reconstructed three-dimensionally (3D) using DeltaViewer software. The 3D-reconstructed image of the sheath of the axon and the CC demonstrated the continuity between the two structures, especially when the image was rotated using DeltaViewer.
Collapse
Affiliation(s)
- Nobuko Katagiri
- 1 Faculty of Nursing, Hirosaki Gakuin University, 20-7 Minori-cho, Hirosaki, Aomori 036-8231, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Xie X, Gilbert M, Petley-Ragan L, Auld VJ. Loss of focal adhesions in glia disrupts both glial and photoreceptor axon migration in the Drosophila visual system. Development 2014; 141:3072-83. [PMID: 25053436 DOI: 10.1242/dev.101972] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Many aspects of glial development are regulated by extracellular signals, including those from the extracellular matrix (ECM). Signals from the ECM are received by cell surface receptors, including the integrin family. Previously, we have shown that Drosophila integrins form adhesion complexes with Integrin-linked kinase and talin in the peripheral nerve glia and have conserved roles in glial sheath formation. However, integrin function in other aspects of glial development is unclear. The Drosophila eye imaginal disc (ED) and optic stalk (OS) complex is an excellent model with which to study glial migration, differentiation and glia-neuron interactions. We studied the roles of the integrin complexes in these glial developmental processes during OS/eye development. The common beta subunit βPS and two alpha subunits, αPS2 and αPS3, are located in puncta at both glia-glia and glia-ECM interfaces. Depletion of βPS integrin and talin by RNAi impaired the migration and distribution of glia within the OS resulting in morphological defects. Reduction of integrin or talin in the glia also disrupted photoreceptor axon outgrowth leading to axon stalling in the OS and ED. The neuronal defects were correlated with a disruption of the carpet glia tube paired with invasion of glia into the core of the OS and the formation of a glial cap. Our results suggest that integrin-mediated extracellular signals are important for multiple aspects of glial development and non-autonomously affect axonal migration during Drosophila eye development.
Collapse
Affiliation(s)
- Xiaojun Xie
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | - Mary Gilbert
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | - Lindsay Petley-Ragan
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | - Vanessa J Auld
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| |
Collapse
|
38
|
Kim SN, Jeibmann A, Halama K, Witte HT, Wälte M, Matzat T, Schillers H, Faber C, Senner V, Paulus W, Klämbt C. ECM stiffness regulates glial migration in Drosophila and mammalian glioma models. Development 2014; 141:3233-42. [DOI: 10.1242/dev.106039] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell migration is an important feature of glial cells. Here, we used the Drosophila eye disc to decipher the molecular network controlling glial migration. We stimulated glial motility by pan-glial PDGF receptor (PVR) activation and identified several genes acting downstream of PVR. Drosophila lox is a non-essential gene encoding a secreted protein that stiffens the extracellular matrix (ECM). Glial-specific knockdown of Integrin results in ECM softening. Moreover, we show that lox expression is regulated by Integrin signaling and vice versa, suggesting that a positive-feedback loop ensures a rigid ECM in the vicinity of migrating cells. The general implication of this model was tested in a mammalian glioma model, where a Lox-specific inhibitor unraveled a clear impact of ECM rigidity in glioma cell migration.
Collapse
Affiliation(s)
- Su Na Kim
- Institute of Neurobiology, University of Münster, Münster 48149, Germany
| | - Astrid Jeibmann
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Kathrin Halama
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Hanna Teresa Witte
- Institute of Neurobiology, University of Münster, Münster 48149, Germany
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Mike Wälte
- Institute of Physiology II, University Hospital Münster, Münster 48149, Germany
| | - Till Matzat
- Institute of Neurobiology, University of Münster, Münster 48149, Germany
| | - Hermann Schillers
- Institute of Physiology II, University Hospital Münster, Münster 48149, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, University Hospital Münster, Münster 48149, Germany
| | - Volker Senner
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Werner Paulus
- Institute of Neuropathology, University Hospital Münster, Münster 48149, Germany
| | - Christian Klämbt
- Institute of Neurobiology, University of Münster, Münster 48149, Germany
| |
Collapse
|
39
|
Meyer S, Schmidt I, Klämbt C. Glia ECM interactions are required to shape the Drosophila nervous system. Mech Dev 2014; 133:105-16. [PMID: 24859129 DOI: 10.1016/j.mod.2014.05.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 05/10/2014] [Accepted: 05/14/2014] [Indexed: 10/25/2022]
Abstract
Organs are characterized by a specific shape that is often remodeled during development. The dynamics of organ shape is in particular evident during the formation of the Drosophila nervous system. During embryonic stages the central nervous system compacts, whereas selective growth occurs during larval stages. The nervous system is covered by a layer of surface glial cells that form the blood brain barrier and a thick extracellular matrix called neural lamella. The size of the neural lamella is dynamically adjusted to the growing nervous system and we show here that perineurial glial cells secrete proteases to remodel this matrix. Moreover, an imbalance in proteolytic activity results in an abnormal shape of the nervous system. To identify further components controlling nervous system shape we performed an RNAi based screen and identified the gene nolo, which encodes an ADAMTS-like protein. We generated loss of function alleles and demonstrate a requirement in glial cells. Mutant nolo larvae, however, do not show an abnormal nervous system shape. The only predicted off-target of the nolo(dsRNA) is Oatp30B, which encodes an organic anion transporting protein characterized by an extracellular protease inhibitor domain. Loss of function mutants were generated and double mutant analyses demonstrate a genetic interaction between nolo and Oatp30B which prevented the generation of maternal zygotic mutant larvae.
Collapse
Affiliation(s)
- Silke Meyer
- Institute of Neurobiology, University of Münster, 48149 Münster, Germany
| | - Imke Schmidt
- Institute of Neurobiology, University of Münster, 48149 Münster, Germany
| | - Christian Klämbt
- Institute of Neurobiology, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
40
|
Ghosh A, Kling T, Snaidero N, Sampaio JL, Shevchenko A, Gras H, Geurten B, Göpfert MC, Schulz JB, Voigt A, Simons M. A global in vivo Drosophila RNAi screen identifies a key role of ceramide phosphoethanolamine for glial ensheathment of axons. PLoS Genet 2013; 9:e1003980. [PMID: 24348263 PMCID: PMC3861124 DOI: 10.1371/journal.pgen.1003980] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/12/2013] [Indexed: 01/08/2023] Open
Abstract
Glia are of vital importance for all complex nervous system. One of the many functions of glia is to insulate and provide trophic and metabolic support to axons. Here, using glial-specific RNAi knockdown in Drosophila, we silenced 6930 conserved genes in adult flies to identify essential genes and pathways. Among our screening hits, metabolic processes were highly represented, and genes involved in carbohydrate and lipid metabolic pathways appeared to be essential in glia. One critical pathway identified was de novo ceramide synthesis. Glial knockdown of lace, a subunit of the serine palmitoyltransferase associated with hereditary sensory and autonomic neuropathies in humans, resulted in ensheathment defects of peripheral nerves in Drosophila. A genetic dissection study combined with shotgun high-resolution mass spectrometry of lipids showed that levels of ceramide phosphoethanolamine are crucial for axonal ensheathment by glia. A detailed morphological and functional analysis demonstrated that the depletion of ceramide phosphoethanolamine resulted in axonal defasciculation, slowed spike propagation, and failure of wrapping glia to enwrap peripheral axons. Supplementing sphingosine into the diet rescued the neuropathy in flies. Thus, our RNAi study in Drosophila identifies a key role of ceramide phosphoethanolamine in wrapping of axons by glia.
Collapse
Affiliation(s)
- Aniket Ghosh
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
- Department of Neurology, University of Göttingen, Göttingen, Germany
| | - Tina Kling
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
- Department of Neurology, University of Göttingen, Göttingen, Germany
| | - Nicolas Snaidero
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
- Department of Neurology, University of Göttingen, Göttingen, Germany
| | - Julio L. Sampaio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrej Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Heribert Gras
- Department of Cellular Neurobiology, University of Göttingen, Göttingen, Germany
| | - Bart Geurten
- Department of Cellular Neurobiology, University of Göttingen, Göttingen, Germany
| | - Martin C. Göpfert
- Department of Cellular Neurobiology, University of Göttingen, Göttingen, Germany
| | - Jörg B. Schulz
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Aaron Voigt
- Department of Neurology, University Medical Center, RWTH Aachen, Aachen, Germany
| | - Mikael Simons
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
- Department of Neurology, University of Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
41
|
von Hilchen CM, Bustos AE, Giangrande A, Technau GM, Altenhein B. Predetermined embryonic glial cells form the distinct glial sheaths of the Drosophila peripheral nervous system. Development 2013; 140:3657-68. [PMID: 23903191 PMCID: PMC3915570 DOI: 10.1242/dev.093245] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
One of the numerous functions of glial cells in Drosophila is the ensheathment of neurons to isolate them from the potassium-rich haemolymph, thereby establishing the blood-brain barrier. Peripheral nerves of flies are surrounded by three distinct glial cell types. Although all embryonic peripheral glia (ePG) have been identified on a single-cell level, their contribution to the three glial sheaths is not known. We used the Flybow system to label and identify each individual ePG in the living embryo and followed them into third instar larva. We demonstrate that all ePG persist until the end of larval development and some even to adulthood. We uncover the origin of all three glial sheaths and describe the larval differentiation of each peripheral glial cell in detail. Interestingly, just one ePG (ePG2) exhibits mitotic activity during larval stages, giving rise to up to 30 glial cells along a single peripheral nerve tract forming the outermost perineurial layer. The unique mitotic ability of ePG2 and the layer affiliation of additional cells were confirmed by in vivo ablation experiments and layer-specific block of cell cycle progression. The number of cells generated by this glial progenitor and hence the control of perineurial hyperplasia correlate with the length of the abdominal nerves. By contrast, the wrapping and subperineurial glia layers show enormous hypertrophy in response to larval growth. This characterisation of the embryonic origin and development of each glial sheath will facilitate functional studies, as they can now be addressed distinctively and genetically manipulated in the embryo.
Collapse
|
42
|
Coutinho-Budd J, Freeman MR. Probing the enigma: unraveling glial cell biology in invertebrates. Curr Opin Neurobiol 2013; 23:1073-9. [PMID: 23896311 DOI: 10.1016/j.conb.2013.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/02/2013] [Accepted: 07/02/2013] [Indexed: 12/11/2022]
Abstract
Despite their predominance in the nervous system, the precise ways in which glial cells develop and contribute to overall neural function remain poorly defined in any organism. Investigations in simple model organisms have identified remarkable morphological, molecular, and functional similarities between invertebrate and vertebrate glial subtypes. Invertebrates like Drosophila and Caenorhabditis elegans offer an abundance of tools for in vivo genetic manipulation of single cells or whole populations of glia, ease of access to neural tissues throughout development, and the opportunity for forward genetic analysis of fundamental aspects of glial cell biology. These features suggest that invertebrate model systems have high potential for vastly improving the understanding of glial biology. This review highlights recent work in Drosophila and other invertebrates that reveal new insights into basic mechanisms involved in glial development.
Collapse
Affiliation(s)
- Jaeda Coutinho-Budd
- Neurobiology Department, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | | |
Collapse
|
43
|
Cell adhesion in Drosophila: versatility of cadherin and integrin complexes during development. Curr Opin Cell Biol 2012; 24:702-12. [PMID: 22938782 DOI: 10.1016/j.ceb.2012.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/16/2012] [Accepted: 07/26/2012] [Indexed: 01/22/2023]
Abstract
We highlight recent progress in understanding cadherin and integrin function in the model organism Drosophila. New functions for these adhesion receptors continue to be discovered in this system, emphasising the importance of cell adhesion within the developing organism and showing that the requirement for cell adhesion changes between cell types. New ways to control adhesion have been discovered, including controlling the expression and recruitment of adhesion components, their posttranslational modification, recycling and turnover. Importantly, even ubiquitous adhesion components can function differently in distinct cellular contexts.
Collapse
|
44
|
Abstract
Kinesin heavy chain (Khc) is crucially required for axonal transport and khc mutants show axonal swellings and paralysis. Here, we demonstrate that in Drosophila khc is equally important in glial cells. Glial-specific downregulation of khc by RNA interference suppresses neuronal excitability and results in spastic flies. The specificity of the phenotype was verified by interspecies rescue experiments and further mutant analyses. Khc is mostly required in the subperineurial glia forming the blood-brain barrier. Following glial-specific knockdown, peripheral nerves are swollen with maldistributed mitochondria. To better understand khc function, we determined Khc-dependent Rab proteins in glia and present evidence that Neurexin IV, a well known blood-brain barrier constituent, is one of the relevant cargo proteins. Our work shows that the role of Khc for neuronal excitability must be considered in the light of its necessity for directed transport in glia.
Collapse
|
45
|
Brink DL, Gilbert M, Xie X, Petley-Ragan L, Auld VJ. Glial processes at the Drosophila larval neuromuscular junction match synaptic growth. PLoS One 2012; 7:e37876. [PMID: 22666403 PMCID: PMC3362601 DOI: 10.1371/journal.pone.0037876] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 04/25/2012] [Indexed: 11/19/2022] Open
Abstract
Glia are integral participants in synaptic physiology, remodeling and maturation from blowflies to humans, yet how glial structure is coordinated with synaptic growth is unknown. To investigate the dynamics of glial development at the Drosophila larval neuromuscular junction (NMJ), we developed a live imaging system to establish the relationship between glia, neuronal boutons, and the muscle subsynaptic reticulum. Using this system we observed processes from two classes of peripheral glia present at the NMJ. Processes from the subperineurial glia formed a blood-nerve barrier around the axon proximal to the first bouton. Processes from the perineurial glial extended beyond the end of the blood-nerve barrier into the NMJ where they contacted synapses and extended across non-synaptic muscle. Growth of the glial processes was coordinated with NMJ growth and synaptic activity. Increasing synaptic size through elevated temperature or the highwire mutation increased the extent of glial processes at the NMJ and conversely blocking synaptic activity and size decreased the presence and size of glial processes. We found that elevated temperature was required during embryogenesis in order to increase glial expansion at the nmj. Therefore, in our live imaging system, glial processes at the NMJ are likely indirectly regulated by synaptic changes to ensure the coordinated growth of all components of the tripartite larval NMJ.
Collapse
Affiliation(s)
- Deidre L. Brink
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mary Gilbert
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiaojun Xie
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lindsay Petley-Ragan
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vanessa J. Auld
- Department of Zoology, Cell and Developmental Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|