1
|
Chang YC, Peng YJ, Lee JY, Chang KT. Peripheral glia and neurons jointly regulate activity-induced synaptic remodeling at the Drosophila neuromuscular junction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600908. [PMID: 39005352 PMCID: PMC11244886 DOI: 10.1101/2024.06.27.600908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In the nervous system, reliable communication depends on the ability of neurons to adaptively remodel their synaptic structure and function in response to changes in neuronal activity. While neurons are the main drivers of synaptic plasticity, glial cells are increasingly recognized for their roles as active modulators. However, the underlying molecular mechanisms remain unclear. Here, using Drosophila neuromuscular junction as a model system for a tripartite synapse, we show that peripheral glial cells collaborate with neurons at the NMJ to regulate activity-induced synaptic remodeling, in part through a protein called shriveled (Shv). Shv is an activator of integrin signaling previously shown to be released by neurons during intense stimulation at the fly NMJ to regulate activity-induced synaptic remodeling. We demonstrate that Shv is also present in peripheral glia, and glial Shv is both necessary and sufficient for synaptic remodeling. However, unlike neuronal Shv, glial Shv does not activate integrin signaling at the NMJ. Instead, it regulates synaptic plasticity in two ways: 1) maintaining the extracellular balance of neuronal Shv proteins to regulate integrin signaling, and 2) controlling ambient extracellular glutamate concentration to regulate postsynaptic glutamate receptor abundance. Loss of glial cells showed the same phenotype as loss of Shv in glia. Together, these results reveal that neurons and glial cells homeostatically regulate extracellular Shv protein levels to control activity-induced synaptic remodeling. Additionally, peripheral glia maintains postsynaptic glutamate receptor abundance and contribute to activity-induced synaptic remodeling by regulating ambient glutamate concentration at the fly NMJ.
Collapse
|
2
|
Petitgas C, Seugnet L, Dulac A, Matassi G, Mteyrek A, Fima R, Strehaiano M, Dagorret J, Chérif-Zahar B, Marie S, Ceballos-Picot I, Birman S. Metabolic and neurobehavioral disturbances induced by purine recycling deficiency in Drosophila. eLife 2024; 12:RP88510. [PMID: 38700995 PMCID: PMC11068357 DOI: 10.7554/elife.88510] [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] [Indexed: 05/05/2024] Open
Abstract
Adenine phosphoribosyltransferase (APRT) and hypoxanthine-guanine phosphoribosyltransferase (HGPRT) are two structurally related enzymes involved in purine recycling in humans. Inherited mutations that suppress HGPRT activity are associated with Lesch-Nyhan disease (LND), a rare X-linked metabolic and neurological disorder in children, characterized by hyperuricemia, dystonia, and compulsive self-injury. To date, no treatment is available for these neurological defects and no animal model recapitulates all symptoms of LND patients. Here, we studied LND-related mechanisms in the fruit fly. By combining enzymatic assays and phylogenetic analysis, we confirm that no HGPRT activity is expressed in Drosophila melanogaster, making the APRT homolog (Aprt) the only purine-recycling enzyme in this organism. Whereas APRT deficiency does not trigger neurological defects in humans, we observed that Drosophila Aprt mutants show both metabolic and neurobehavioral disturbances, including increased uric acid levels, locomotor impairments, sleep alterations, seizure-like behavior, reduced lifespan, and reduction of adenosine signaling and content. Locomotor defects could be rescued by Aprt re-expression in neurons and reproduced by knocking down Aprt selectively in the protocerebral anterior medial (PAM) dopaminergic neurons, the mushroom bodies, or glia subsets. Ingestion of allopurinol rescued uric acid levels in Aprt-deficient mutants but not neurological defects, as is the case in LND patients, while feeding adenosine or N6-methyladenosine (m6A) during development fully rescued the epileptic behavior. Intriguingly, pan-neuronal expression of an LND-associated mutant form of human HGPRT (I42T), but not the wild-type enzyme, resulted in early locomotor defects and seizure in flies, similar to Aprt deficiency. Overall, our results suggest that Drosophila could be used in different ways to better understand LND and seek a cure for this dramatic disease.
Collapse
Affiliation(s)
- Céline Petitgas
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
- Metabolomic and Proteomic Biochemistry Laboratory, Necker-Enfants Malades Hospital and Paris Cité UniversityParisFrance
| | - Laurent Seugnet
- Integrated Physiology of the Brain Arousal Systems (WAKING), Lyon Neuroscience Research Centre, INSERM/CNRS/UCBL1BronFrance
| | - Amina Dulac
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Giorgio Matassi
- Dipartimento di Scienze Agroalimentari, Ambientali e Animali, University of UdineUdineItaly
- UMR “Ecology and Dynamics of Anthropogenic Systems” (EDYSAN), CNRS, Université de Picardie Jules VerneAmiensFrance
| | - Ali Mteyrek
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Rebecca Fima
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Marion Strehaiano
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Joana Dagorret
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Baya Chérif-Zahar
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| | - Sandrine Marie
- Laboratory of Metabolic Diseases, Cliniques Universitaires Saint-Luc, Université catholique de LouvainBrusselsBelgium
| | - Irène Ceballos-Picot
- Metabolomic and Proteomic Biochemistry Laboratory, Necker-Enfants Malades Hospital and Paris Cité UniversityParisFrance
| | - Serge Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research UniversityParisFrance
| |
Collapse
|
3
|
Miao H, Wei Y, Lee SG, Wu Z, Kaur J, Kim WJ. Glia-specific expression of neuropeptide receptor Lgr4 regulates development and adult physiology in Drosophila. J Neurosci Res 2024; 102:e25271. [PMID: 38284837 DOI: 10.1002/jnr.25271] [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: 02/08/2023] [Revised: 10/10/2023] [Accepted: 10/28/2023] [Indexed: 01/30/2024]
Abstract
Similar to the human brain, Drosophila glia may well be divided into several subtypes that each carries out specific functions. Glial GPCRs play key roles in crosstalk between neurons and glia. Drosophila Lgr4 (dLgr4) is a human relaxin receptor homolog involved in angiogenesis, cardiovascular regulation, collagen remodeling, and wound healing. A recent study suggests that ilp7 might be the ligand for Lgr4 and regulates escape behavior of Drosophila larvae. Here we demonstrate that Drosophila Lgr4 expression in glial cells, not neurons, is necessary for early development, adult behavior, and lifespan. Reducing the Lgr4 level in glial cells disrupts Drosophila development, while knocking down other LGR family members in glia has no impact. Adult-specific knockdown of Lgr4 in glia but not neurons reduce locomotion, male reproductive success, and animal longevity. The investigation of how glial expression of Lgr4 contributes to this behavioral alteration will increase our understanding of how insulin signaling via glia selectively modulates neuronal activity and behavior.
Collapse
Affiliation(s)
- Hongyu Miao
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
| | - Yanan Wei
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
| | - Seung Gee Lee
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Zekun Wu
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
| | - Jasdeep Kaur
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Woo Jae Kim
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Bharat V, Durairaj AS, Vanhauwaert R, Li L, Muir CM, Chandra S, Kwak CS, Le Guen Y, Nandakishore P, Hsieh CH, Rensi SE, Altman RB, Greicius MD, Feng L, Wang X. A mitochondrial inside-out iron-calcium signal reveals drug targets for Parkinson's disease. Cell Rep 2023; 42:113544. [PMID: 38060381 PMCID: PMC10804639 DOI: 10.1016/j.celrep.2023.113544] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/11/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023] Open
Abstract
Dysregulated iron or Ca2+ homeostasis has been reported in Parkinson's disease (PD) models. Here, we discover a connection between these two metals at the mitochondria. Elevation of iron levels causes inward mitochondrial Ca2+ overflow, through an interaction of Fe2+ with mitochondrial calcium uniporter (MCU). In PD neurons, iron accumulation-triggered Ca2+ influx across the mitochondrial surface leads to spatially confined Ca2+ elevation at the outer mitochondrial membrane, which is subsequently sensed by Miro1, a Ca2+-binding protein. A Miro1 blood test distinguishes PD patients from controls and responds to drug treatment. Miro1-based drug screens in PD cells discover Food and Drug Administration-approved T-type Ca2+-channel blockers. Human genetic analysis reveals enrichment of rare variants in T-type Ca2+-channel subtypes associated with PD status. Our results identify a molecular mechanism in PD pathophysiology and drug targets and candidates coupled with a convenient stratification method.
Collapse
Affiliation(s)
- Vinita Bharat
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aarooran S Durairaj
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Roeland Vanhauwaert
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Li Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Colin M Muir
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Graduate Program of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sujyoti Chandra
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chulhwan S Kwak
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Institut du Cerveau - Paris Brain Institute - ICM, 75013 Paris, France
| | | | - Chung-Han Hsieh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stefano E Rensi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liang Feng
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinnan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
5
|
Bhatnagar A, Parmar V, Barbieri N, Bearoff F, Elefant F, Kortagere S. Novel EAAT2 activators improve motor and cognitive impairment in a transgenic model of Huntington's disease. Front Behav Neurosci 2023; 17:1176777. [PMID: 37351153 PMCID: PMC10282606 DOI: 10.3389/fnbeh.2023.1176777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Glutamate excitotoxicity is causal in striatal neurodegeneration underlying motor dysfunction and cognitive deficits in Huntington's disease (HD). Excitatory amino acid transporter 2 (EAAT2), the predominant glutamate transporter accounting for >90% of glutamate transport, plays a key role in preventing excitotoxicity by clearing excess glutamate from the intrasynaptic cleft. Accordingly, EAAT2 has emerged as a promising therapeutic target for prevention of neuronal excitotoxicity underlying HD and other neurodegenerative diseases. Methods We have previously designed novel EAAT2 positive allosteric modulator GT951, GTS467, and GTS551, with low nanomolar efficacy in glutamate uptake and favorable pharmacokinetic properties. In this study, we test the neuroprotective abilities of these novel EAAT2 activators in vivo using the robust Drosophila HD transgenic model expressing human huntingtin gene with expanded repeats (Htt128Q). Results All three compounds significantly restored motor function impaired under HD pathology over a wide dose range. Additionally, treatment with all three compounds significantly improved HD-associated olfactory associative learning and short-term memory defects, while GT951 and GTS551 also improved middle-term memory in low-performing group. Similarly, treatment with GT951 and GTS551 partially protected against early mortality observed in our HD model. Further, treatment with all three EAAT2 activators induced epigenetic expression of EAAT2 Drosophila homolog and several cognition-associated genes. Conclusion Together, these results highlight the efficacy of GT951, GTS467 and GTS551 in treating motor and cognitive impairments under HD pathology and support their development for treatment of HD.
Collapse
Affiliation(s)
- Akanksha Bhatnagar
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
| | - Visha Parmar
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Nicholas Barbieri
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Frank Bearoff
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Felice Elefant
- Department of Biology, Papadakis Integrated Sciences Building, Drexel University, Philadelphia, PA, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
6
|
Scott H, Novikov B, Ugur B, Allen B, Mertsalov I, Monagas-Valentin P, Koff M, Baas Robinson S, Aoki K, Veizaj R, Lefeber DJ, Tiemeyer M, Bellen H, Panin V. Glia-neuron coupling via a bipartite sialylation pathway promotes neural transmission and stress tolerance in Drosophila. eLife 2023; 12:e78280. [PMID: 36946697 PMCID: PMC10110239 DOI: 10.7554/elife.78280] [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: 03/01/2022] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Modification by sialylated glycans can affect protein functions, underlying mechanisms that control animal development and physiology. Sialylation relies on a dedicated pathway involving evolutionarily conserved enzymes, including CMP-sialic acid synthetase (CSAS) and sialyltransferase (SiaT) that mediate the activation of sialic acid and its transfer onto glycan termini, respectively. In Drosophila, CSAS and DSiaT genes function in the nervous system, affecting neural transmission and excitability. We found that these genes function in different cells: the function of CSAS is restricted to glia, while DSiaT functions in neurons. This partition of the sialylation pathway allows for regulation of neural functions via a glia-mediated control of neural sialylation. The sialylation genes were shown to be required for tolerance to heat and oxidative stress and for maintenance of the normal level of voltage-gated sodium channels. Our results uncovered a unique bipartite sialylation pathway that mediates glia-neuron coupling and regulates neural excitability and stress tolerance.
Collapse
Affiliation(s)
- Hilary Scott
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Boris Novikov
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Berrak Ugur
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Brooke Allen
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Ilya Mertsalov
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Pedro Monagas-Valentin
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Melissa Koff
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| | - Sarah Baas Robinson
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Raisa Veizaj
- Translational Metabolic Laboratory, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenNetherlands
| | - Dirk J Lefeber
- Translational Metabolic Laboratory, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenNetherlands
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of GeorgiaAthensUnited States
| | - Hugo Bellen
- Departments of Molecular and Human Genetics and Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Vladislav Panin
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege StationUnited States
| |
Collapse
|
7
|
Mocci I, Casu MA, Sogos V, Liscia A, Angius R, Cadeddu F, Fanti M, Muroni P, Talani G, Diana A, Collu M, Setzu MD. Effects of memantine on mania-like phenotypes exhibited by Drosophila Shaker mutants. CNS Neurosci Ther 2023. [PMID: 36942502 DOI: 10.1111/cns.14145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/23/2023] Open
Abstract
INTRODUCTION Increased glutamate levels and electrolytic fluctuations have been observed in acutely manic patients. Despite some efficacy of the non-competitive NMDA receptor antagonist memantine (Mem), such as antidepressant-like and mood-stabilizer drugs in clinical studies, its specific mechanisms of action are still uncertain. The present study aims to better characterize the Drosophila melanogaster fly Shaker mutants (SH), as a translational model of manic episodes within bipolar disorder in humans, and to investigate the potential anti-manic properties of Mem. METHODS AND RESULTS Our findings showed typical behavioral abnormalities in SH, which mirrored with the overexpression of NMDAR-NR1 protein subunit, matched well to glutamate up-regulation. Such molecular features were associated to a significant reduction of SH brain volume in comparison to Wild Type strain flies (WT). Here we report on the ability of Mem treatment to ameliorate behavioral aberrations of SH (similar to that of Lithium), and its ability to reduce NMDAR-NR1 over-expression. CONCLUSIONS Our results show the involvement of the glutamatergic system in the SH, given the interaction between the Shaker channel and the NMDA receptor, suggesting this model as a promising tool for studying the neurobiology of bipolar disorders. Moreover, our results show Mem as a potential disease-modifying therapy, providing insight on new mechanisms of action.
Collapse
Affiliation(s)
- Ignazia Mocci
- Institute of Translational Pharmacology, National Research Council, Science and Technology Park of Sardinia, Cagliari, Italy
| | - Maria Antonietta Casu
- Institute of Translational Pharmacology, National Research Council, Science and Technology Park of Sardinia, Cagliari, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Anna Liscia
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Rossella Angius
- Unit of Biomedical Research Support, NMR Laboratory and Bioanalytical Technologies, Sardegna Ricerche, Science and Technology Park of Sardinia, Cagliari, Italy
| | - Francesca Cadeddu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maura Fanti
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Patrizia Muroni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Giuseppe Talani
- Institute of Neuroscience, National Research Council, Monserrato, Italy
| | - Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Collu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Dolores Setzu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| |
Collapse
|
8
|
Rahmani Z, Surabhi S, Rojo-Cortés F, Dulac A, Jenny A, Birman S. Lamp1 Deficiency Enhances Sensitivity to α-Synuclein and Oxidative Stress in Drosophila Models of Parkinson Disease. Int J Mol Sci 2022; 23:13078. [PMID: 36361864 PMCID: PMC9657416 DOI: 10.3390/ijms232113078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/28/2022] [Accepted: 10/19/2022] [Indexed: 11/15/2023] Open
Abstract
Parkinson disease (PD) is a common neurodegenerative condition affecting people predominantly at old age that is characterized by a progressive loss of midbrain dopaminergic neurons and by the accumulation of α-synuclein-containing intraneuronal inclusions known as Lewy bodies. Defects in cellular degradation processes such as the autophagy-lysosomal pathway are suspected to be involved in PD progression. The mammalian Lysosomal-associated membrane proteins LAMP1 and LAMP2 are transmembrane glycoproteins localized in lysosomes and late endosomes that are involved in autophagosome/lysosome maturation and function. Here, we show that the lack of Drosophila Lamp1, the homolog of LAMP1 and LAMP2, severely increased fly susceptibility to paraquat, a pro-oxidant compound known as a potential PD inducer in humans. Moreover, the loss of Lamp1 also exacerbated the progressive locomotor defects induced by the expression of PD-associated mutant α-synuclein A30P (α-synA30P) in dopaminergic neurons. Remarkably, the ubiquitous re-expression of Lamp1 in a mutant context fully suppressed all these defects and conferred significant resistance towards both PD factors above that of wild-type flies. Immunostaining analysis showed that the brain levels of α-synA30P were unexpectedly decreased in young adult Lamp1-deficient flies expressing this protein in comparison to non-mutant controls. This suggests that Lamp1 could neutralize α-synuclein toxicity by promoting the formation of non-pathogenic aggregates in neurons. Overall, our findings reveal a novel role for Drosophila Lamp1 in protecting against oxidative stress and α-synuclein neurotoxicity in PD models, thus furthering our understanding of the function of its mammalian homologs.
Collapse
Affiliation(s)
- Zohra Rahmani
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, UMR 8249, CNRS, ESPCI Paris, PSL University, 75005 Paris, France
| | - Satya Surabhi
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Francisca Rojo-Cortés
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, UMR 8249, CNRS, ESPCI Paris, PSL University, 75005 Paris, France
| | - Amina Dulac
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, UMR 8249, CNRS, ESPCI Paris, PSL University, 75005 Paris, France
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Serge Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, UMR 8249, CNRS, ESPCI Paris, PSL University, 75005 Paris, France
| |
Collapse
|
9
|
De Backer JF, Grunwald Kadow IC. A role for glia in cellular and systemic metabolism: insights from the fly. CURRENT OPINION IN INSECT SCIENCE 2022; 53:100947. [PMID: 35772690 DOI: 10.1016/j.cois.2022.100947] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
Excitability and synaptic transmission make neurons high-energy consumers. However, neurons do not store carbohydrates or lipids. Instead, they need support cells to fuel their metabolic demands. This role is assumed by glia, both in vertebrates and invertebrates. Many questions remain regarding the coupling between neuronal activity and energy demand on the one hand, and nutrient supply by glia on the other hand. Here, we review recent advances showing that fly glia, similar to their role in vertebrates, fuel neurons in times of high energetic demand, such as during memory formation and long-term storage. Vertebrate glia also play a role in the modulation of neurons, their communication, and behavior, including food search and feeding. We discuss recent literature pointing to similar roles of fly glia in behavior and metabolism.
Collapse
Affiliation(s)
- Jean-François De Backer
- Technical University of Munich, School of Life Sciences, Liesel-Beckmann-Str. 4, 85354 Freising, Germany; University of Bonn, Faculty of Medicine, UKB, Institute of Physiology II, Nussallee 11, 53115 Bonn, Germany
| | - Ilona C Grunwald Kadow
- Technical University of Munich, School of Life Sciences, Liesel-Beckmann-Str. 4, 85354 Freising, Germany; University of Bonn, Faculty of Medicine, UKB, Institute of Physiology II, Nussallee 11, 53115 Bonn, Germany.
| |
Collapse
|
10
|
Chen X, Li J, Gao Z, Yang Y, Kuang W, Dong Y, Chua GH, Huang X, Jiang B, Tian H, Wang Y, Huang X, Li Y, Lam SM, Shui G. Endogenous ceramide phosphoethanolamine modulates circadian rhythm via neural-glial coupling in Drosophila. Natl Sci Rev 2022; 9:nwac148. [PMID: 36713590 PMCID: PMC9875363 DOI: 10.1093/nsr/nwac148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 06/08/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
While endogenous lipids are known to exhibit rhythmic oscillations, less is known about how specific lipids modulate circadian behavior. Through a series of loss-of-function and gain-of-function experiments on ceramide phosphoethanolamine (CPE) synthase of Drosophila, we demonstrated that pan-glial-specific deficiency in membrane CPE, the structural analog of mammalian sphingomyelin (SM), leads to arrhythmic locomotor behavior and shortens lifespan, while the reverse is true for increasing CPE. Comparative proteomics uncovered dysregulated synaptic glutamate utilization and transport in CPE-deficient flies. An extensive genetic screen was conducted to verify the role of differentially expressed proteins in circadian regulation. Arrhythmic locomotion under cpes1 mutant background was rescued only by restoring endogenous CPE or SM through expressing their respective synthases. Our results underscore the essential role of CPE in maintaining synaptic glutamate homeostasis and modulating circadian behavior in Drosophila. The findings suggest that region-specific elevations of functional membrane lipids can benefit circadian regulation.
Collapse
Affiliation(s)
| | | | - Zhongbao Gao
- University of Chinese Academy of Sciences, Beijing 100049, China,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yang Yang
- University of Chinese Academy of Sciences, Beijing 100049, China,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wenqing Kuang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yue Dong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gek Huey Chua
- LipidALL Technologies Company Limited, Changzhou213022, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Binhua Jiang
- LipidALL Technologies Company Limited, Changzhou213022, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Li
- University of Chinese Academy of Sciences, Beijing 100049, China,State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | | | |
Collapse
|
11
|
Dravecz N, Shaw T, Davies I, Brown C, Ormerod L, Vu G, Walker T, Taank T, Shirras AD, Broughton SJ. Reduced Insulin Signaling Targeted to Serotonergic Neurons but Not Other Neuronal Subtypes Extends Lifespan in Drosophila melanogaster. Front Aging Neurosci 2022; 14:893444. [PMID: 35865744 PMCID: PMC9294736 DOI: 10.3389/fnagi.2022.893444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Reduced Insulin/IGF-like signaling (IIS) plays an evolutionarily conserved role in improving longevity and some measures of health-span in model organisms. Recent studies, however, have found a disconnection between lifespan extension and behavioral health-span. We have previously shown that reduction of IIS in Drosophila neurons extends female lifespan but does not improve negative geotaxis senescence and has a detrimental effect on exploratory walking senescence in both sexes. We hypothesize that individual neuronal subtypes respond differently to IIS changes, thus the behavioral outcomes of pan-neuronal IIS reduction are the balance of positive, negative and neutral functional effects. In order to further understand how reduced IIS in neurons independently modulates lifespan and locomotor behavioral senescence we expressed a dominant negative Insulin receptor transgene selectively in individual neuronal subtypes and measured the effects on lifespan and two measures of locomotor senescence, negative geotaxis and exploratory walking. IIS reduction in cholinergic, GABAergic, dopaminergic, glutamatergic, and octopaminergic neurons was found to have either no affect or a detrimental effect on lifespan and locomotor senescence. However, reduction of IIS selectively in serotonergic neurons resulted in extension of lifespan in females with no effect on locomotor senescence. These data indicate that individual neuronal subtypes respond differently to IIS changes in the modulation of lifespan and locomotor senescence, and identify a specific role for the insulin receptor in serotonergic neurons in the modulation of lifespan.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Susan J. Broughton
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| |
Collapse
|
12
|
Huang Q, Liao C, Ge F, Ao J, Liu T. Acetylcholine bidirectionally regulates learning and memory. JOURNAL OF NEURORESTORATOLOGY 2022. [DOI: 10.1016/j.jnrt.2022.100002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
13
|
Peng H, Guo D, Shan W, Liu Z, Wang H, Ma L, Xu B, Guo X. Identification of the AccCDK1 gene in Apis cerana cerana and its relationship with the oxidative stress response. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 182:105048. [PMID: 35249658 DOI: 10.1016/j.pestbp.2022.105048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/16/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
The cyclin-dependent kinase (CDK) protein family plays an important role in regulating life functions, such as the cell cycle and metabolism. This study reports the first cloning and functional analysis of A. cerana cerana CDK1 (AccCDK1). The distribution profile of AccCDK1 in different developmental periods and different tissues was determined. The experimental results showed that the distribution of AccCDK1 was tissue-specific. AccCDK1 distribution at the transcriptional and translational levels was affected by stress conditions induced by H2O2, UV, HgCl2, CdCl2, extreme temperatures (4 °C, 44 °C) and pesticides (avermectin, lambda-cyhalothrin, haloxyfop-R-methyl, and glyphosate), which resulted in changes in the expression levels. These results suggest that AccCDK1 may have an important part to play in honey bee resistance to stress. The expression of a recombinant AccCDK1 protein in vitro enhanced the antistress capacities of E. coli and yeast, which suggests that AccCDK1 is related to the stress response. When AccCDK1 was silenced, the expression of some antioxidant genes was downregulated, and the enzymatic potencies of superoxide dismutase (SOD), peroxidase (POD) and catalase (CAT) were reduced, which suggests that AccCDK1 takes part in the body's resistance to oxidative stress upon external stimulation by influencing relevant antioxidants. Notably, the survival rate of A. cerana cerana under high-temperature-induced stress decreased after AccCDK1 silencing, which verifies our results. In conclusion, we found that AccCDK1 played an indispensable function in resisting oxidative stress and maintaining normal cellular functions.
Collapse
Affiliation(s)
- Hongyan Peng
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Dezheng Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Wenlu Shan
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Zhenguo Liu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Hongfang Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Lanting Ma
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| |
Collapse
|
14
|
Fernandes J, Varghese J. Sexually dimorphic microRNA miR-190 regulates lifespan in male Drosophila. RNA Biol 2022; 19:1085-1093. [PMID: 36178157 PMCID: PMC9542781 DOI: 10.1080/15476286.2022.2127544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
microRNAs are short noncoding RNAs that buffer fluctuations in gene expression in a myriad of physiological conditions. Here, we carried out a screen to identify the role of microRNAs in the maintenance of age-dependent neuronal functions in adult Drosophila. We report that miR-190 acts in the neurons to regulate lifespan, neuronal maintanence and age-related locomotor activity specifically in male flies. miR-190, a highly conserved microRNA, shows higher expression levels in male flies. Our data suggest that miR-190 functions by regulating target genes that are involved in maintaining neuronal activity and lifespan in male flies.
Collapse
Affiliation(s)
- Jervis Fernandes
- School of Biology, Indian Institute of Science Education and Research (IISER TVM), Thiruvananthapuram, India
| | - Jishy Varghese
- School of Biology, Indian Institute of Science Education and Research (IISER TVM), Thiruvananthapuram, India
| |
Collapse
|
15
|
Li X, Chien C, Han Y, Sun Z, Chen X, Dickman D. Autocrine inhibition by a glutamate-gated chloride channel mediates presynaptic homeostatic depression. SCIENCE ADVANCES 2021; 7:eabj1215. [PMID: 34851664 PMCID: PMC8635443 DOI: 10.1126/sciadv.abj1215] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/14/2021] [Indexed: 06/13/2023]
Abstract
Homeostatic modulation of presynaptic neurotransmitter release is a fundamental form of plasticity that stabilizes neural activity, where presynaptic homeostatic depression (PHD) can adaptively diminish synaptic strength. PHD has been proposed to operate through an autocrine mechanism to homeostatically depress release probability in response to excess glutamate release at the Drosophila neuromuscular junction. This model implies the existence of a presynaptic glutamate autoreceptor. We systematically screened all neuronal glutamate receptors in the fly genome and identified the glutamate-gated chloride channel (GluClα) to be required for the expression of PHD. Pharmacological, genetic, and Ca2+ imaging experiments demonstrate that GluClα acts locally at axonal terminals to drive PHD. Unexpectedly, GluClα localizes and traffics with synaptic vesicles to drive presynaptic inhibition through an activity-dependent anionic conductance. Thus, GluClα operates as both a sensor and effector of PHD to adaptively depress neurotransmitter release through an elegant autocrine inhibitory signaling mechanism at presynaptic terminals.
Collapse
Affiliation(s)
- Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
- USC Neuroscience Graduate Program, Los Angeles, CA, 90089, USA
| | - Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
- USC Neuroscience Graduate Program, Los Angeles, CA, 90089, USA
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
- USC Neuroscience Graduate Program, Los Angeles, CA, 90089, USA
| | - Zihan Sun
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Xun Chen
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
- USC Neuroscience Graduate Program, Los Angeles, CA, 90089, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| |
Collapse
|
16
|
Ramnarine TJS, Grath S, Parsch J. Natural variation in the transcriptional response of Drosophila melanogaster to oxidative stress. G3-GENES GENOMES GENETICS 2021; 12:6409858. [PMID: 34747443 PMCID: PMC8727983 DOI: 10.1093/g3journal/jkab366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/15/2021] [Indexed: 11/26/2022]
Abstract
Broadly distributed species must cope with diverse and changing environmental conditions, including various forms of stress. Cosmopolitan populations of Drosophila melanogaster are more tolerant to oxidative stress than those from the species’ ancestral range in sub-Saharan Africa, and the degree of tolerance is associated with an insertion/deletion polymorphism in the 3′ untranslated region of the Metallothionein A (MtnA) gene that varies clinally in frequency. We examined oxidative stress tolerance and the transcriptional response to oxidative stress in cosmopolitan and sub-Saharan African populations of D. melanogaster, including paired samples with allelic differences at the MtnA locus. We found that the effect of the MtnA polymorphism on oxidative stress tolerance was dependent on the genomic background, with the deletion allele increasing tolerance only in a northern, temperate population. Genes that were differentially expressed under oxidative stress included MtnA and other metallothioneins, as well as those involved in glutathione metabolism and other genes known to be part of the oxidative stress response or the general stress response. A gene coexpression analysis revealed further genes and pathways that respond to oxidative stress including those involved in additional metabolic processes, autophagy, and apoptosis. There was a significant overlap among the genes induced by oxidative and cold stress, which suggests a shared response pathway to these two stresses. Interestingly, the MtnA deletion was associated with consistent changes in the expression of many genes across all genomic backgrounds, regardless of the expression level of the MtnA gene itself. We hypothesize that this is an indirect effect driven by the loss of microRNA binding sites within the MtnA 3′ untranslated region.
Collapse
Affiliation(s)
- Timothy J S Ramnarine
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität (LMU) München, Planegg-Martinsried 82152, Germany
| | - Sonja Grath
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität (LMU) München, Planegg-Martinsried 82152, Germany
| | - John Parsch
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität (LMU) München, Planegg-Martinsried 82152, Germany
| |
Collapse
|
17
|
Li L, Conradson DM, Bharat V, Kim MJ, Hsieh CH, Minhas PS, Papakyrikos AM, Durairaj AS, Ludlam A, Andreasson KI, Partridge L, Cianfrocco MA, Wang X. A mitochondrial membrane-bridging machinery mediates signal transduction of intramitochondrial oxidation. Nat Metab 2021; 3:1242-1258. [PMID: 34504353 PMCID: PMC8460615 DOI: 10.1038/s42255-021-00443-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Mitochondria are the main site for generating reactive oxygen species, which are key players in diverse biological processes. However, the molecular pathways of redox signal transduction from the matrix to the cytosol are poorly defined. Here we report an inside-out redox signal of mitochondria. Cysteine oxidation of MIC60, an inner mitochondrial membrane protein, triggers the formation of disulfide bonds and the physical association of MIC60 with Miro, an outer mitochondrial membrane protein. The oxidative structural change of this membrane-crossing complex ultimately elicits cellular responses that delay mitophagy, impair cellular respiration and cause oxidative stress. Blocking the MIC60-Miro interaction or reducing either protein, genetically or pharmacologically, extends lifespan and health-span of healthy fruit flies, and benefits multiple models of Parkinson's disease and Friedreich's ataxia. Our discovery provides a molecular basis for common treatment strategies against oxidative stress.
Collapse
Affiliation(s)
- Li Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Devon M Conradson
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Vinita Bharat
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Min Joo Kim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Chung-Han Hsieh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Paras S Minhas
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Neurosciences Intradepartmental Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Amanda M Papakyrikos
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Graduate Program in Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Anthony Ludlam
- Life Sciences Institute & Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Katrin I Andreasson
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| | - Linda Partridge
- Institute of Healthy Ageing, Genetics, Evolution and Environment, University College London, London, UK
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Michael A Cianfrocco
- Life Sciences Institute & Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Xinnan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Maternal & Child Health Research Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
18
|
Liu Y, Zhu F, Shen Z, Moural TW, Liu L, Li Z, Liu X, Xu H. Glutaredoxins and thioredoxin peroxidase involved in defense of emamectin benzoate induced oxidative stress in Grapholita molesta. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 176:104881. [PMID: 34119223 DOI: 10.1016/j.pestbp.2021.104881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/08/2021] [Accepted: 05/19/2021] [Indexed: 06/12/2023]
Abstract
Glutaredoxins (Grxs) and thioredoxin peroxidases (Tpxs) are major antioxidant enzyme families involved in regulating cellular redox homeostasis and in defense of enhanced oxidative stress through scavenging reactive oxygen species (ROS). However, the functions of these enzymes have not been reported in the oriental fruit moth, Grapholita molesta (Busck), a worldwide pest of stone and pome fruits. Here, we identified four new antioxidant genes, GmGrx, GmGrx3, GmGrx5, and GmTpx which were induced by exposure with emamectin benzoate, a commonly used biopesticide for G. molesta control. Other environmental factors (low and high temperatures, Escherichia coli and Metarhizium anisopliae) also significantly induced the expression of these genes. After GmGrx or GmTpx silenced by RNA interference (RNAi), the percentage of larval survival to emamectin benzoate were significantly decreased, demonstrating that GmGrx and GmTpx are involved in protecting G. molesta from stresses induced by emamectin benzoate. Furthermore, silenced GmGrx, GmGrx3, GmGrx5, or GmTpx significantly enhanced the enzymatic activities of superoxide dismutase (SOD) (except GmTpx) and peroxidase (POD), as well as the contents of hydrogen peroxide and metabolites ascorbate. Taken together, our results suggest that GmGrx, GmGrx3, GmGrx5, and GmTpx may play critical roles in antioxidant defense. Specially, GmGrx and GmTpx contribute to the defense of oxidative damage induced by exposure to emamectin benzoate through scavenging excessive ROS in G. molesta. Our findings provided a theoretical basis for understanding functions of insect glutaredoxin and peroxidase systems.
Collapse
Affiliation(s)
- Yanjun Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China; Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Fang Zhu
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Zhongjian Shen
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Timothy W Moural
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Lining Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zhen Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xiaoxia Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Huanli Xu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China.
| |
Collapse
|
19
|
Meng J, Wang L, Wang C, Zhao G, Wang H, Xu B, Guo X. AccPDIA6 from Apis cerana cerana plays important roles in antioxidation. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 175:104830. [PMID: 33993956 DOI: 10.1016/j.pestbp.2021.104830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/06/2021] [Accepted: 03/13/2021] [Indexed: 06/12/2023]
Abstract
PDIA6 is a member of the protein disulfide isomerase (PDI) family, shows disulfide isomerase activity and oxidoreductase activity, and can act as a molecular chaperone. Its biological functions include modulating apoptosis, regulating the proliferation and invasion of cancer cells, supporting thrombosis and modulating insulin secretion. However, the roles of PDIA6 in Apis cerana cerana are poorly understood. Herein, we obtained the PDIA6 gene from A. cerana cerana (AccPDIA6). We investigated the expression patterns of AccPDIA6 in response to oxidative stress induced by H2O2, UV, HgCl2, extreme temperatures (4 °C, 42 °C) and pesticides (thiamethoxam and hexythiazox) and found that AccPDIA6 was upregulated by these treatments. Western blot analysis indicated that AccPDIA6 was also upregulated by oxidative stress at the protein level. In addition, a survival test demonstrated that the survival rate of E. coli cells expressing AccPDIA6 increased under oxidative stress, suggesting a possible antioxidant function of AccPDIA6. In addition, we tested the transcripts of other antioxidant genes and found that some of them were downregulated in AccPDIA6 knockdown samples. It was also discovered that the antioxidant enzymatic activity of superoxide dismutase (SOD) decreased in AccPDIA6-silenced bees. Moreover, the survival rate of AccPDIA6 knockdown bees decreased under oxidative stress, implying that AccPDIA6 may function in the oxidative stress response by enhancing the viability of honeybees. Taken together, these results indicated that AccPDIA6 may play an essential role in counteracting oxidative stress.
Collapse
Affiliation(s)
- Jie Meng
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Lijun Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Chen Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Guangdong Zhao
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Hongfang Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Taian, Shandong 271018, PR China.
| |
Collapse
|
20
|
Bozzo M, Lacalli TC, Obino V, Caicci F, Marcenaro E, Bachetti T, Manni L, Pestarino M, Schubert M, Candiani S. Amphioxus neuroglia: Molecular characterization and evidence for early compartmentalization of the developing nerve cord. Glia 2021; 69:1654-1678. [PMID: 33624886 DOI: 10.1002/glia.23982] [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: 11/25/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
Glial cells play important roles in the development and homeostasis of metazoan nervous systems. However, while their involvement in the development and function in the central nervous system (CNS) of vertebrates is increasingly well understood, much less is known about invertebrate glia and the evolutionary history of glial cells more generally. An investigation into amphioxus glia is therefore timely, as this organism is the best living proxy for the last common ancestor of all chordates, and hence provides a window into the role of glial cell development and function at the transition of invertebrates and vertebrates. We report here our findings on amphioxus glia as characterized by molecular probes correlated with anatomical data at the transmission electron microscopy (TEM) level. The results show that amphioxus glial lineages express genes typical of vertebrate astroglia and radial glia, and that they segregate early in development, forming what appears to be a spatially separate cell proliferation zone positioned laterally, between the dorsal and ventral zones of neural cell proliferation. Our study provides strong evidence for the presence of vertebrate-type glial cells in amphioxus, while highlighting the role played by segregated progenitor cell pools in CNS development. There are implications also for our understanding of glial cells in a broader evolutionary context, and insights into patterns of precursor cell deployment in the chordate nerve cord.
Collapse
Affiliation(s)
- Matteo Bozzo
- Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| | - Thurston C Lacalli
- Biology Department, University of Victoria, Victoria, British Columbia, Canada
| | - Valentina Obino
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Emanuela Marcenaro
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Tiziana Bachetti
- Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| | - Lucia Manni
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Mario Pestarino
- Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| | - Michael Schubert
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer, Villefranche-sur-Mer, France
| | - Simona Candiani
- Department of Earth, Environment and Life Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
21
|
Zhou D, Stobdan T, Visk D, Xue J, Haddad GG. Genetic interactions regulate hypoxia tolerance conferred by activating Notch in excitatory amino acid transporter 1-positive glial cells in Drosophila melanogaster. G3 (BETHESDA, MD.) 2021; 11:jkab038. [PMID: 33576765 PMCID: PMC8022968 DOI: 10.1093/g3journal/jkab038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 01/28/2021] [Indexed: 12/26/2022]
Abstract
Hypoxia is a critical pathological element in many human diseases, including ischemic stroke, myocardial infarction, and solid tumors. Of particular significance and interest of ours are the cellular and molecular mechanisms that underlie susceptibility or tolerance to low O2. Previous studies have demonstrated that Notch signaling pathway regulates hypoxia tolerance in both Drosophila melanogaster and humans. However, the mechanisms mediating Notch-conferred hypoxia tolerance are largely unknown. In this study, we delineate the evolutionarily conserved mechanisms underlying this hypoxia tolerant phenotype. We determined the role of a group of conserved genes that were obtained from a comparative genomic analysis of hypoxia-tolerant D.melanogaster populations and human highlanders living at the high-altitude regions of the world (Tibetans, Ethiopians, and Andeans). We developed a novel dual-UAS/Gal4 system that allows us to activate Notch signaling in the Eaat1-positive glial cells, which remarkably enhances hypoxia tolerance in D.melanogaster, and, simultaneously, knock down a candidate gene in the same set of glial cells. Using this system, we discovered that the interactions between Notch signaling and bnl (fibroblast growth factor), croc (forkhead transcription factor C), or Mkk4 (mitogen-activated protein kinase kinase 4) are important for hypoxia tolerance, at least in part, through regulating neuronal development and survival under hypoxic conditions. Becausethese genetic mechanisms are evolutionarily conserved, this group of genes may serve as novel targets for developing therapeutic strategies and have a strong potential to be translated to humans to treat/prevent hypoxia-related diseases.
Collapse
Affiliation(s)
- Dan Zhou
- Division of Respiratory Medicine, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Tsering Stobdan
- Division of Respiratory Medicine, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - DeeAnn Visk
- Division of Respiratory Medicine, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Jin Xue
- Division of Respiratory Medicine, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Gabriel G Haddad
- Department of Neurosciences, University of California at San Diego, La Jolla, CA 92093, USA
- Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
22
|
Nagai J, Yu X, Papouin T, Cheong E, Freeman MR, Monk KR, Hastings MH, Haydon PG, Rowitch D, Shaham S, Khakh BS. Behaviorally consequential astrocytic regulation of neural circuits. Neuron 2020; 109:576-596. [PMID: 33385325 DOI: 10.1016/j.neuron.2020.12.008] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
Astrocytes are a large and diverse population of morphologically complex cells that exist throughout nervous systems of multiple species. Progress over the last two decades has shown that astrocytes mediate developmental, physiological, and pathological processes. However, a long-standing open question is how astrocytes regulate neural circuits in ways that are behaviorally consequential. In this regard, we summarize recent studies using Caenorhabditis elegans, Drosophila melanogaster, Danio rerio, and Mus musculus. The data reveal diverse astrocyte mechanisms operating in seconds or much longer timescales within neural circuits and shaping multiple behavioral outputs. We also refer to human diseases that have a known primary astrocytic basis. We suggest that including astrocytes in mechanistic, theoretical, and computational studies of neural circuits provides new perspectives to understand behavior, its regulation, and its disease-related manifestations.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; RIKEN Center for Brain Science, 2-1 Hirosawa Wako City, Saitama 351-0198, Japan
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, 514 Burrill Hall, 407 S. Goodwin Ave, Urbana, IL 61801, USA
| | - Thomas Papouin
- Department of Neuroscience, Washington University in St. Louis, School of Medicine, Campus Box 8108, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Marc R Freeman
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Kelly R Monk
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael H Hastings
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - David Rowitch
- Department of Paediatrics, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Departments of Pediatrics and Neurosurgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA.
| |
Collapse
|
23
|
Sarkar S, Murphy MA, Dammer EB, Olsen AL, Rangaraju S, Fraenkel E, Feany MB. Comparative proteomic analysis highlights metabolic dysfunction in α-synucleinopathy. NPJ PARKINSONS DISEASE 2020; 6:40. [PMID: 33311497 PMCID: PMC7732845 DOI: 10.1038/s41531-020-00143-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/12/2020] [Indexed: 12/15/2022]
Abstract
The synaptic protein α-synuclein is linked through genetics and neuropathology to the pathogenesis of Parkinson’s disease and related disorders. However, the mechanisms by which α-synuclein influences disease onset and progression are incompletely understood. To identify pathogenic pathways and therapeutic targets we performed proteomic analysis in a highly penetrant new Drosophila model of α-synucleinopathy. We identified 476 significantly upregulated and 563 significantly downregulated proteins in heads from α-synucleinopathy model flies compared to controls. We then used multiple complementary analyses to identify and prioritize genes and pathways within the large set of differentially expressed proteins for functional studies. We performed Gene Ontology enrichment analysis, integrated our proteomic changes with human Parkinson’s disease genetic studies, and compared the α-synucleinopathy proteome with that of tauopathy model flies, which are relevant to Alzheimer’s disease and related disorders. These approaches identified GTP cyclohydrolase (GCH1) and folate metabolism as candidate mediators of α-synuclein neurotoxicity. In functional validation studies, we found that the knockdown of Drosophila Gch1 enhanced locomotor deficits in α-synuclein transgenic flies, while folate supplementation protected from α-synuclein toxicity. Our integrative analysis suggested that mitochondrial dysfunction was a common downstream mediator of neurodegeneration. Accordingly, Gch1 knockdown enhanced metabolic dysfunction in α-synuclein transgenic fly brains while folate supplementation partially normalized brain bioenergetics. Here we outline and implement an integrative approach to identify and validate potential therapeutic pathways using comparative proteomics and genetics and capitalizing on the facile genetic and pharmacological tools available in Drosophila.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A Murphy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric B Dammer
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Spen modulates lipid droplet content in adult Drosophila glial cells and protects against paraquat toxicity. Sci Rep 2020; 10:20023. [PMID: 33208773 PMCID: PMC7674452 DOI: 10.1038/s41598-020-76891-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/16/2020] [Indexed: 12/21/2022] Open
Abstract
Glial cells are early sensors of neuronal injury and can store lipids in lipid droplets under oxidative stress conditions. Here, we investigated the functions of the RNA-binding protein, SPEN/SHARP, in the context of Parkinson’s disease (PD). Using a data-mining approach, we found that SPEN/SHARP is one of many astrocyte-expressed genes that are significantly differentially expressed in the substantia nigra of PD patients compared with control subjects. Interestingly, the differentially expressed genes are enriched in lipid metabolism-associated genes. In a Drosophila model of PD, we observed that flies carrying a loss-of-function allele of the ortholog split-ends (spen) or with glial cell-specific, but not neuronal-specific, spen knockdown were more sensitive to paraquat intoxication, indicating a protective role for Spen in glial cells. We also found that Spen is a positive regulator of Notch signaling in adult Drosophila glial cells. Moreover, Spen was required to limit abnormal accumulation of lipid droplets in glial cells in a manner independent of its regulation of Notch signaling. Taken together, our results demonstrate that Spen regulates lipid metabolism and storage in glial cells and contributes to glial cell-mediated neuroprotection.
Collapse
|
25
|
Woodling NS, Rajasingam A, Minkley LJ, Rizzo A, Partridge L. Independent glial subtypes delay development and extend healthy lifespan upon reduced insulin-PI3K signalling. BMC Biol 2020; 18:124. [PMID: 32928209 PMCID: PMC7490873 DOI: 10.1186/s12915-020-00854-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/21/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The increasing age of global populations highlights the urgent need to understand the biological underpinnings of ageing. To this end, inhibition of the insulin/insulin-like signalling (IIS) pathway can extend healthy lifespan in diverse animal species, but with trade-offs including delayed development. It is possible that distinct cell types underlie effects on development and ageing; cell-type-specific strategies could therefore potentially avoid negative trade-offs when targeting diseases of ageing, including prevalent neurodegenerative diseases. The highly conserved diversity of neuronal and non-neuronal (glial) cell types in the Drosophila nervous system makes it an attractive system to address this possibility. We have thus investigated whether IIS in distinct glial cell populations differentially modulates development and lifespan in Drosophila. RESULTS We report here that glia-specific IIS inhibition, using several genetic means, delays development while extending healthy lifespan. The effects on lifespan can be recapitulated by adult-onset IIS inhibition, whereas developmental IIS inhibition is dispensable for modulation of lifespan. Notably, the effects we observe on both lifespan and development act through the PI3K branch of the IIS pathway and are dependent on the transcription factor FOXO. Finally, IIS inhibition in several glial subtypes can delay development without extending lifespan, whereas the same manipulations in astrocyte-like glia alone are sufficient to extend lifespan without altering developmental timing. CONCLUSIONS These findings reveal a role for distinct glial subpopulations in the organism-wide modulation of development and lifespan, with IIS in astrocyte-like glia contributing to lifespan modulation but not to developmental timing. Our results enable a more complete picture of the cell-type-specific effects of the IIS network, a pathway whose evolutionary conservation in humans make it tractable for therapeutic interventions. Our findings therefore underscore the necessity for cell-type-specific strategies to optimise interventions for the diseases of ageing.
Collapse
Affiliation(s)
- Nathaniel S Woodling
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Arjunan Rajasingam
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Lucy J Minkley
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Alberto Rizzo
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK
| | - Linda Partridge
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK.
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931, Cologne, Germany.
| |
Collapse
|
26
|
Kim T, Song B, Lee IS. Drosophila Glia: Models for Human Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2020; 21:E4859. [PMID: 32660023 PMCID: PMC7402321 DOI: 10.3390/ijms21144859] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/27/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Glial cells are key players in the proper formation and maintenance of the nervous system, thus contributing to neuronal health and disease in humans. However, little is known about the molecular pathways that govern glia-neuron communications in the diseased brain. Drosophila provides a useful in vivo model to explore the conserved molecular details of glial cell biology and their contributions to brain function and disease susceptibility. Herein, we review recent studies that explore glial functions in normal neuronal development, along with Drosophila models that seek to identify the pathological implications of glial defects in the context of various central nervous system disorders.
Collapse
Affiliation(s)
| | | | - Im-Soon Lee
- Department of Biological Sciences, Center for CHANS, Konkuk University, Seoul 05029, Korea; (T.K.); (B.S.)
| |
Collapse
|
27
|
Yon M, Decoville M, Sarou-Kanian V, Fayon F, Birman S. Spatially-resolved metabolic profiling of living Drosophila in neurodegenerative conditions using 1H magic angle spinning NMR. Sci Rep 2020; 10:9516. [PMID: 32528106 PMCID: PMC7289880 DOI: 10.1038/s41598-020-66218-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Drosophila flies are versatile animal models for the study of gene mutations in neuronal pathologies. Their small size allows performing in vivo Magic Angle Spinning (MAS) experiments to obtain high-resolution 1H nuclear magnetic resonance (NMR) spectra. Here, we use spatially-resolved 1H high-resolution MAS NMR to investigate in vivo metabolite contents in different segments of the fly body. A comparative study of metabolic changes was performed for three neurodegenerative disorders: two cell-specific neuronal and glial models of Huntington disease (HD) and a model of glutamate excitotoxicity. It is shown that these pathologies are characterized by specific and sometimes anatomically localized variations in metabolite concentrations. In two cases, the modifications of 1H MAS NMR spectra localized in fly heads were significant enough to allow the creation of a predictive model.
Collapse
Affiliation(s)
- Maxime Yon
- CEMHTI UPR3079, CNRS, Université d'Orléans, F-45071, Orléans, France
| | | | | | - Franck Fayon
- CEMHTI UPR3079, CNRS, Université d'Orléans, F-45071, Orléans, France
| | - Serge Birman
- GCRN-LPC UMR8249, CNRS, ESPCI Paris, PSL Research University, F-75005, Paris, France
| |
Collapse
|
28
|
Hernandez E, MacNamee SE, Kaplan LR, Lance K, Garcia-Verdugo HD, Farhadi DS, Deer C, Lee SW, Oland LA. The astrocyte network in the ventral nerve cord neuropil of the Drosophila third-instar larva. J Comp Neurol 2020; 528:1683-1703. [PMID: 31909826 DOI: 10.1002/cne.24852] [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: 05/28/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 11/10/2022]
Abstract
Understanding neuronal function at the local and circuit level requires understanding astrocyte function. We have provided a detailed analysis of astrocyte morphology and territory in the Drosophila third-instar ventral nerve cord where there already exists considerable understanding of the neuronal network. Astrocyte shape varies more than previously reported; many have bilaterally symmetrical partners, many have a high percentage of their arborization in adjacent segments, and many have branches that follow structural features. Taken together, our data are consistent with, but not fully explained by, a model of a developmental growth process dominated by competitive or repulsive interactions between astrocytes. Our data suggest that the model should also include cell-autonomous aspects, as well as the use of structural features for growth. Variation in location of arborization territory for identified astrocytes was great enough that a standardized scheme of neuropil division among the six astrocytes that populate each hemi-segment is not possible at the third instar. The arborizations of the astrocytes can extend across neuronal functional domains. The ventral astrocyte in particular, whose territory can extend well into the proprioceptive region of the neuropil, has no obvious branching pattern that correlates with domains of particular sensory modalities, suggesting that the astrocyte would respond to neuronal activity in any of the sensory modalities, perhaps integrating across them. This study sets the stage for future studies that will generate a robust, functionally oriented connectome that includes both partners in neuronal circuits-the neurons and the glial cells, providing the foundation necessary for studies to elucidate neuron-glia interactions in this neuropil.
Collapse
Key Words
- RRID:Abcam Cat# ab6953, RRID:AB_955010
- RRID:BDSC Cat# 30125, RRID:BDSC_30125
- RRID:BDSC Cat# 38760, RRID:BDSC_38760
- RRID:BDSC Cat# 4775, RRID:BDSC_4775
- RRID:BDSC Cat# 5692, RRID:BDSC_5692
- RRID:BDSC Cat# 64085, RRID:BDSC_64085
- RRID:BDSC Cat# 6938, RRID:BDSC_6938
- RRID:Bio-rad Cat # MCA1360, RRID:AB_322378
- RRID:Cell Signaling Technology Cat # 3724, RRID:AB_1549585
- RRID:DSHB Cat# 1D4, RRID:AB_528235
- RRID:DSHB Cat# nc82, RRID:AB_2314866
- RRID:Jackson ImmunoResearch Labs Cat# 115-167-003, RRID:AB_2338709
- RRID:Molecular Probes Cat# 6455, RRID:AB_2314543
- RRID:Molecular Probes Cat# A-21236, RRID:AB_141725
- RRID:Novus Cat # NBP1-06712, RRID:AB_1625981
- RRID:Thermo Fisher Scientific Cat# A-11034, RRID:AB_2576217.
- glial cells
- neuron-glia interaction
Collapse
Affiliation(s)
- Ernesto Hernandez
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,University of Illinois at Chicago School of Medicine, Rockford, Illinois
| | - Sarah E MacNamee
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,Inscopix, Palo Alto, California
| | - Leah R Kaplan
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,Consortium for Science, Policy & Outcomes, Arizona State University, Washington, DC, Washington
| | - Kim Lance
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| | | | - Dara S Farhadi
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona
| | - Christine Deer
- Department of Neuroscience, University of Arizona, Tucson, Arizona.,Research Technologies Group, Data Visualization Team, University of Arizona, University Information Technology Service, Tucson, Arizona
| | - Si W Lee
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| | - Lynne A Oland
- Department of Neuroscience, University of Arizona, Tucson, Arizona
| |
Collapse
|
29
|
Li X, Goel P, Wondolowski J, Paluch J, Dickman D. A Glutamate Homeostat Controls the Presynaptic Inhibition of Neurotransmitter Release. Cell Rep 2019; 23:1716-1727. [PMID: 29742428 PMCID: PMC5973541 DOI: 10.1016/j.celrep.2018.03.130] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/24/2018] [Accepted: 03/28/2018] [Indexed: 02/07/2023] Open
Abstract
We have interrogated the synaptic dialog that enables the bi-directional, homeostatic control of pre-synaptic efficacy at the glutamatergic Drosophila neuromuscular junction (NMJ). We find that homeo-static depression and potentiation use disparate genetic, induction, and expression mechanisms. Specifically, homeostatic potentiation is achieved through reduced CaMKII activity postsynaptically and increased abundance of active zone material presynaptically at one of the two neuronal subtypes innervating the NMJ, while homeostatic depression occurs without alterations in CaMKII activity and is expressed at both neuronal subtypes. Furthermore, homeostatic depression is only induced through excess presynaptic glutamate release and operates with disregard to the postsynaptic response. We propose that two independent homeostats modulate presynaptic efficacy at the Drosophila NMJ: one is an intercellular signaling system that potentiates synaptic strength following diminished postsynaptic excitability, while the other adaptively modulates presynaptic glutamate release through an autocrine mechanism without feedback from the postsynaptic compartment.
Collapse
Affiliation(s)
- Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, CA; USC Neuroscience Graduate Program, University of Southern California, Los Angeles, CA
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA; USC Graduate Program in Molecular and Computational Biology, University of Southern California, Los Angeles, CA
| | - Joyce Wondolowski
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| | - Jeremy Paluch
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA.
| |
Collapse
|
30
|
Hsieh CH, Li L, Vanhauwaert R, Nguyen KT, Davis MD, Bu G, Wszolek ZK, Wang X. Miro1 Marks Parkinson's Disease Subset and Miro1 Reducer Rescues Neuron Loss in Parkinson's Models. Cell Metab 2019; 30:1131-1140.e7. [PMID: 31564441 PMCID: PMC6893131 DOI: 10.1016/j.cmet.2019.08.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/07/2019] [Accepted: 08/28/2019] [Indexed: 12/31/2022]
Abstract
The identification of molecular targets and pharmacodynamic markers for Parkinson's disease (PD) will empower more effective clinical management and experimental therapies. Miro1 is localized on the mitochondrial surface and mediates mitochondrial motility. Miro1 is removed from depolarized mitochondria to facilitate their clearance via mitophagy. Here, we explore the clinical utility of Miro1 for detecting PD and for gauging potential treatments. We measure the Miro1 response to mitochondrial depolarization using biochemical assays in skin fibroblasts from a broad spectrum of PD patients and discover that more than 94% of the patients' fibroblast cell lines fail to remove Miro1 following depolarization. We identify a small molecule that can repair this defect of Miro1 in PD fibroblasts. Treating patient-derived neurons and fly models with this compound rescues the locomotor deficits and dopaminergic neurodegeneration. Our results indicate that tracking this Miro1 marker and engaging in Miro1-based therapies could open new avenues to personalized medicine.
Collapse
Affiliation(s)
- Chung-Han Hsieh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Li Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Roeland Vanhauwaert
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Mary D Davis
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Xinnan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Olsen AL, Feany MB. Glial α-synuclein promotes neurodegeneration characterized by a distinct transcriptional program in vivo. Glia 2019; 67:1933-1957. [PMID: 31267577 DOI: 10.1002/glia.23671] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/29/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022]
Abstract
α-Synucleinopathies are neurodegenerative diseases that are characterized pathologically by α-synuclein inclusions in neurons and glia. The pathologic contribution of glial α-synuclein in these diseases is not well understood. Glial α-synuclein may be of particular importance in multiple system atrophy (MSA), which is defined pathologically by glial cytoplasmic α-synuclein inclusions. We have previously described Drosophila models of neuronal α-synucleinopathy, which recapitulate key features of the human disorders. We have now expanded our model to express human α-synuclein in glia. We demonstrate that expression of α-synuclein in glia alone results in α-synuclein aggregation, death of dopaminergic neurons, impaired locomotor function, and autonomic dysfunction. Furthermore, co-expression of α-synuclein in both neurons and glia worsens these phenotypes as compared to expression of α-synuclein in neurons alone. We identify unique transcriptomic signatures induced by glial as opposed to neuronal α-synuclein. These results suggest that glial α-synuclein may contribute to the burden of pathology in the α-synucleinopathies through a cell type-specific transcriptional program. This new Drosophila model system enables further mechanistic studies dissecting the contribution of glial and neuronal α-synuclein in vivo, potentially shedding light on mechanisms of disease that are especially relevant in MSA but also the α-synucleinopathies more broadly.
Collapse
Affiliation(s)
- Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
Knockdown of the neuronal gene Lim3 at the early stages of development affects mitochondrial function and lifespan in Drosophila. Mech Ageing Dev 2019; 181:29-41. [DOI: 10.1016/j.mad.2019.111121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/29/2019] [Accepted: 05/30/2019] [Indexed: 01/08/2023]
|
33
|
Transcriptional Regulation of the Glutamate/GABA/Glutamine Cycle in Adult Glia Controls Motor Activity and Seizures in Drosophila. J Neurosci 2019; 39:5269-5283. [PMID: 31064860 PMCID: PMC6607755 DOI: 10.1523/jneurosci.1833-18.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 01/23/2023] Open
Abstract
The fruitfly Drosophila melanogaster has been extensively used as a genetic model for the maintenance of nervous system's functions. Glial cells are of utmost importance in regulating the neuronal functions in the adult organism and in the progression of neurological pathologies. Through a microRNA-based screen in adult Drosophila glia, we uncovered the essential role of a major glia developmental determinant, repo, in the adult fly. Here, we report that Repo expression is continuously required in adult glia to transcriptionally regulate the highly conserved function of neurotransmitter recycling in both males and females. Transient loss of Repo dramatically shortens fly lifespan, triggers motor deficits, and increases the sensibility to seizures, partly due to the impairment of the glutamate/GABA/glutamine cycle. Our findings highlight the pivotal role of transcriptional regulation of genes involved in the glutamate/GABA/glutamine cycle in glia to control neurotransmitter levels in neurons and their behavioral output. The mechanism identified here in Drosophila exemplifies how adult functions can be modulated at the transcriptional level and suggest an active synchronized regulation of genes involved in the same pathway. The process of neurotransmitter recycling is of essential importance in human epileptic and psychiatric disorders and our findings may thus have important consequences for the understanding of the role that transcriptional regulation of neurotransmitter recycling in astrocytes has in human disease. SIGNIFICANCE STATEMENT Glial cells are an essential support to neurons in adult life and have been involved in a number of neurological disorders. What controls the maintenance and modulation of glial functions in adult life is not fully characterized. Through a miR overexpression screen in adult glia in Drosophila, we identify an essential role in adult glia of repo, which directs glial differentiation during embryonic development. Repo levels modulate, via transcriptional regulation, the ability of glial cells to support neurons in the glutamate/GABA/glutamine cycle. This leads to significant abnormalities in motor behavior as assessed through a novel automated paradigm. Our work points to the importance of transcriptional regulation in adult glia for neurotransmitter recycling, a key process in several human neurological disorders.
Collapse
|
34
|
Matsuno M, Horiuchi J, Ofusa K, Masuda T, Saitoe M. Inhibiting Glutamate Activity during Consolidation Suppresses Age-Related Long-Term Memory Impairment in Drosophila. iScience 2019; 15:55-65. [PMID: 31030182 PMCID: PMC6487374 DOI: 10.1016/j.isci.2019.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/24/2018] [Accepted: 04/08/2019] [Indexed: 01/17/2023] Open
Abstract
In Drosophila, long-term memory (LTM) formation requires increases in glial gene expression. Klingon (Klg), a cell adhesion molecule expressed in both neurons and glia, induces expression of the glial transcription factor, Repo. However, glial signaling downstream of Repo has been unclear. Here we demonstrate that Repo increases expression of the glutamate transporter, EAAT1, and EAAT1 is required during consolidation of LTM. The expressions of Klg, Repo, and EAAT1 decrease upon aging, suggesting that age-related impairments in LTM are caused by dysfunction of the Klg-Repo-EAAT1 pathway. Supporting this idea, overexpression of Repo or EAAT1 rescues age-associated impairments in LTM. Pharmacological inhibition of glutamate activity during consolidation improves LTM in klg mutants and aged flies. Altogether, our results indicate that LTM formation requires glial-dependent inhibition of glutamate signaling during memory consolidation, and aging disrupts this process by inhibiting the Klg-Repo-EAAT1 pathway.
Collapse
Affiliation(s)
- Motomi Matsuno
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8502, Japan
| | - Junjiro Horiuchi
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8502, Japan
| | - Kyoko Ofusa
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8502, Japan
| | - Tomoko Masuda
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8502, Japan
| | - Minoru Saitoe
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8502, Japan.
| |
Collapse
|
35
|
Cusumano P, Damulewicz M, Carbognin E, Caccin L, Puricella A, Specchia V, Bozzetti MP, Costa R, Mazzotta GM. The RNA Helicase BELLE Is Involved in Circadian Rhythmicity and in Transposons Regulation in Drosophila melanogaster. Front Physiol 2019; 10:133. [PMID: 30842743 PMCID: PMC6392097 DOI: 10.3389/fphys.2019.00133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/04/2019] [Indexed: 02/05/2023] Open
Abstract
Circadian clocks control and synchronize biological rhythms of several behavioral and physiological phenomena in most, if not all, organisms. Rhythm generation relies on molecular auto-regulatory oscillations of interlocked transcriptional-translational feedback loops. Rhythmic clock-gene expression is at the base of rhythmic protein accumulation, though post-transcriptional and post-translational mechanisms have evolved to adjust and consolidate the proper pace of the clock. In Drosophila, BELLE, a conserved DEAD-box RNA helicase playing important roles in reproductive capacity, is involved in the small RNA-mediated regulation associated to the piRNA pathway. Here, we report that BELLE is implicated in the circadian rhythmicity and in the regulation of endogenous transposable elements (TEs) in both nervous system and gonads. We suggest that BELLE acts as important element in the piRNA-mediated regulation of the TEs and raise the hypothesis that this specific regulation could represent another level of post-transcriptional control adopted by the clock to ensure the proper rhythmicity.
Collapse
Affiliation(s)
- Paola Cusumano
- Department of Biology, University of Padua, Padua, Italy
| | - Milena Damulewicz
- Department of Cell Biology and Imaging, Jagiellonian University, Kraków, Poland
| | | | - Laura Caccin
- Department of Biology, University of Padua, Padua, Italy
| | - Antonietta Puricella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Valeria Specchia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Maria Pia Bozzetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Rodolfo Costa
- Department of Biology, University of Padua, Padua, Italy
| | | |
Collapse
|
36
|
Abstract
As the nervous system evolved from the diffused to centralised form, the neurones were joined by the appearance of the supportive cells, the neuroglia. Arguably, these non-neuronal cells evolve into a more diversified cell family than the neurones are. The first ancestral neuroglia appeared in flatworms being mesenchymal in origin. In the nematode C. elegans proto-astrocytes/supportive glia of ectodermal origin emerged, albeit the ensheathment of axons by glial cells occurred later in prawns. The multilayered myelin occurred by convergent evolution of oligodendrocytes and Schwann cells in vertebrates above the jawless fishes. Nutritive partitioning of the brain from the rest of the body appeared in insects when the hemolymph-brain barrier, a predecessor of the blood-brain barrier was formed. The defensive cellular mechanism required specialisation of bona fide immune cells, microglia, a process that occurred in the nervous system of leeches, bivalves, snails, insects and above. In ascending phylogeny, new type of glial cells, such as scaffolding radial glia, appeared and as the bran sizes enlarged, the glia to neurone ratio increased. Humans possess some unique glial cells not seen in other animals.
Collapse
|
37
|
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
|
38
|
Shen ZJ, Liu YJ, Gao XH, Liu XM, Zhang SD, Li Z, Zhang QW, Liu XX. Molecular Identification of Two Thioredoxin Genes From Grapholita molesta and Their Function in Resistance to Emamectin Benzoate. Front Physiol 2018; 9:1421. [PMID: 30410444 PMCID: PMC6210739 DOI: 10.3389/fphys.2018.01421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/18/2018] [Indexed: 01/28/2023] Open
Abstract
Thioredoxins (Trxs), a member of the thioredoxin system, play crucial roles in maintaining intracellular redox homeostasis and protecting organisms against oxidative stress. In this study, we cloned and characterized two genes, GmTrx2 and GmTrx-like1, from Grapholita molesta. Sequence analysis showed that GmTrx2 and GmTrx-like1 had highly conserved active sites CGPC and CXXC motif, respectively, and shared high sequence identity with selected insect species. The quantitative real-time polymerase chain reaction results revealed that GmTrx2 was mainly detected at first instar, whereas GmTrx-like1 was highly concentrated at prepupa day. The transcripts of GmTrx2 and GmTrx-like1 were both highly expressed in the head and salivary glands. The expression levels of GmTrx2 and GmTrx-like1 were induced by low or high temperature, E. coli, M. anisopliae, H2O2, and pesticides (emamectin benzoate). We further detected interference efficiency of GmTrx2 and GmTrx-like1 in G. molesta larvae and found that peroxidase capacity, hydrogen peroxide content, and ascorbate content all increased after knockdown of GmTrx2 or GmTrx-like1. Furthermore, the hydrogen peroxide concentration was increased by emamectin benzoate and the sensitivity for larvae to emamectin benzoate was improved after GmTrx2 or GmTrx-like1 was silenced. Our results indicated that GmTrx2 and GmTrx-like1 played vital roles in protecting G. molesta against oxidative damage and also provided the theoretical basis for understanding the antioxidant defense mechanisms of the Trx system in insects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiao-Xia Liu
- Department of Entomology, MOA Key Laboratory of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
39
|
Focal adhesion molecules regulate astrocyte morphology and glutamate transporters to suppress seizure-like behavior. Proc Natl Acad Sci U S A 2018; 115:11316-11321. [PMID: 30327343 DOI: 10.1073/pnas.1800830115] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Astrocytes are important regulators of neural circuit function and behavior in the healthy and diseased nervous system. We screened for molecules in Drosophila astrocytes that modulate neuronal hyperexcitability and identified multiple components of focal adhesion complexes (FAs). Depletion of astrocytic Tensin, β-integrin, Talin, focal adhesion kinase (FAK), or matrix metalloproteinase 1 (Mmp1), resulted in enhanced behavioral recovery from genetic or pharmacologically induced seizure. Overexpression of Mmp1, predicted to activate FA signaling, led to a reciprocal enhancement of seizure severity. Blockade of FA-signaling molecules in astrocytes at basal levels of CNS excitability resulted in reduced astrocytic coverage of the synaptic neuropil and expression of the excitatory amino acid transporter EAAT1. However, induction of hyperexcitability after depletion of FA-signaling components resulted in enhanced astrocyte coverage and an approximately twofold increase in EAAT1 levels. Our work identifies FA-signaling molecules as important regulators of astrocyte outgrowth and EAAT1 expression under normal physiological conditions. Paradoxically, in the context of hyperexcitability, this pathway negatively regulates astrocytic process outgrowth and EAAT1 expression, and their blockade leading to enhanced recovery from seizure.
Collapse
|
40
|
Wang L, Wang C, Li H, Yang X, Wang Y, Guo X, Xu B. Isolation of
AccGalectin1
from
Apis cerana cerana
and its functions in development and adverse stress response. J Cell Biochem 2018; 120:671-684. [DOI: 10.1002/jcb.27424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/12/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Lijun Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University Taian Shandong China
| | - Chen Wang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University Taian Shandong China
| | - Han Li
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University Taian Shandong China
| | - Xinxin Yang
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University Taian Shandong China
| | - Ying Wang
- College of Animal Science and Technology, Shandong Agricultural University Taian Shandong China
| | - Xingqi Guo
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University Taian Shandong China
| | - Baohua Xu
- College of Animal Science and Technology, Shandong Agricultural University Taian Shandong China
| |
Collapse
|
41
|
Ren Q, Awasaki T, Wang YC, Huang YF, Lee T. Lineage-guided Notch-dependent gliogenesis by Drosophila multi-potent progenitors. Development 2018; 145:dev.160127. [PMID: 29764857 DOI: 10.1242/dev.160127] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/08/2018] [Indexed: 12/27/2022]
Abstract
Macroglial cells in the central nervous system exhibit regional specialization and carry out region-specific functions. Diverse glial cells arise from specific progenitors in specific spatiotemporal patterns. This raises an interesting possibility that glial precursors with distinct developmental fates exist that govern region-specific gliogenesis. Here, we have mapped the glial progeny produced by the Drosophila type II neuroblasts, which, like vertebrate radial glia cells, yield both neurons and glia via intermediate neural progenitors (INPs). Distinct type II neuroblasts produce different characteristic sets of glia. A single INP can make both astrocyte-like and ensheathing glia, which co-occupy a relatively restrictive subdomain. Blocking apoptosis uncovers further lineage distinctions in the specification, proliferation and survival of glial precursors. Both the switch from neurogenesis to gliogenesis and the subsequent glial expansion depend on Notch signaling. Taken together, lineage origins preconfigure the development of individual glial precursors with involvement of serial Notch actions in promoting gliogenesis.
Collapse
Affiliation(s)
- Qingzhong Ren
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Takeshi Awasaki
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Yu-Chun Wang
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Yu-Fen Huang
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Tzumin Lee
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| |
Collapse
|
42
|
Kounatidis I, Chtarbanova S. Role of Glial Immunity in Lifespan Determination: A Drosophila Perspective. Front Immunol 2018; 9:1362. [PMID: 29942319 PMCID: PMC6004738 DOI: 10.3389/fimmu.2018.01362] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/01/2018] [Indexed: 11/13/2022] Open
Abstract
Increasing body of evidence indicates that proper glial function plays an important role in neuroprotection and in organismal physiology throughout lifespan. Work done in the model organism Drosophila melanogaster has revealed important aspects of glial cell biology in the contexts of longevity and neurodegeneration. In this mini review, we summarize recent findings from work done in the fruit fly Drosophila about the role of glia in maintaining a healthy status during animal’s life and discuss the involvement of glial innate immune pathways in lifespan and neurodegeneration. Overactive nuclear factor kappa B (NF-κB) pathways and defective phagocytosis appear to be major contributors to lifespan shortening and neuropathology. Glial NF-κB silencing on the other hand, extends lifespan possibly through an immune–neuroendocrine axis. Given the evolutionary conservation of NF-κB innate immune signaling and of macrophage ontogeny across fruit flies, rodents, and humans, the above observations in glia could potentially support efforts for therapeutic interventions targeting to ameliorate age-related pathologies.
Collapse
Affiliation(s)
- Ilias Kounatidis
- Cell Biology, Development, and Genetics Laboratory, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
43
|
Sakakibara Y, Sekiya M, Fujisaki N, Quan X, Iijima KM. Knockdown of wfs1, a fly homolog of Wolfram syndrome 1, in the nervous system increases susceptibility to age- and stress-induced neuronal dysfunction and degeneration in Drosophila. PLoS Genet 2018; 14:e1007196. [PMID: 29357349 PMCID: PMC5794194 DOI: 10.1371/journal.pgen.1007196] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/01/2018] [Accepted: 01/09/2018] [Indexed: 01/08/2023] Open
Abstract
Wolfram syndrome (WS), caused by loss-of-function mutations in the Wolfram syndrome 1 gene (WFS1), is characterized by juvenile-onset diabetes mellitus, bilateral optic atrophy, and a wide spectrum of neurological and psychiatric manifestations. WFS1 encodes an endoplasmic reticulum (ER)-resident transmembrane protein, and mutations in this gene lead to pancreatic β-cell death induced by high levels of ER stress. However, the mechanisms underlying neurodegeneration caused by WFS1 deficiency remain elusive. Here, we investigated the role of WFS1 in the maintenance of neuronal integrity in vivo by knocking down the expression of wfs1, the Drosophila homolog of WFS1, in the central nervous system. Neuronal knockdown of wfs1 caused age-dependent behavioral deficits and neurodegeneration in the fly brain. Knockdown of wfs1 in neurons and glial cells resulted in premature death and significantly exacerbated behavioral deficits in flies, suggesting that wfs1 has important functions in both cell types. Although wfs1 knockdown alone did not promote ER stress, it increased the susceptibility to oxidative stress-, excitotoxicity- or tauopathy-induced behavioral deficits, and neurodegeneration. The glutamate release inhibitor riluzole significantly suppressed premature death phenotypes induced by neuronal and glial knockdown of wfs1. This study highlights the protective role of wfs1 against age-associated neurodegeneration and furthers our understanding of potential disease-modifying factors that determine susceptibility and resilience to age-associated neurodegenerative diseases. Wolfram syndrome (WS), a neurodegenerative disorder with an autosomal recessive inheritance pattern, has a variable clinical presentation that includes diabetes mellitus, optic atrophy, and a wide spectrum of neurological and psychiatric manifestations. Homozygous mutations in WFS1 are causative for WS. The prognosis of WS is poor, and most patients die prematurely with respiratory failure due to brain stem atrophy. However, the mechanisms underlying the neurological manifestations of WS remain elusive. In this study, we used the fruit fly Drosophila to examine the neurological features of WS by generating genetically modified flies harboring knockdown of wfs1, the fly homolog of WFS1, in the central nervous system. These flies developed age-dependent behavioral deficits, neurodegeneration and premature death. wfs1-deficient flies were vulnerable to various age-related stressors such as oxidative stress and excitotoxicity, and to neurodegeneration caused by Alzheimer’s disease-related toxic proteins. The premature death phenotype in wfs1-deficient flies was ameliorated by administration of riluzole, which inhibits glutamate-induced excitotoxicity. This study provides insight into the mechanisms underlying neurodegeneration not only in WS, but also in age-associated neurodegenerative diseases such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Yasufumi Sakakibara
- Department of Alzheimer’s Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Michiko Sekiya
- Department of Alzheimer’s Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Naoki Fujisaki
- Department of Alzheimer’s Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3–1 Tanabe-dori, Mizuho-ku, Nagoya, Japan
| | - Xiuming Quan
- Department of Alzheimer’s Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Koichi M. Iijima
- Department of Alzheimer’s Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3–1 Tanabe-dori, Mizuho-ku, Nagoya, Japan
- * E-mail:
| |
Collapse
|
44
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
45
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 964] [Impact Index Per Article: 160.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
46
|
Moore BD, Martin J, de Mena L, Sanchez J, Cruz PE, Ceballos-Diaz C, Ladd TB, Ran Y, Levites Y, Kukar TL, Kurian JJ, McKenna R, Koo EH, Borchelt DR, Janus C, Rincon-Limas D, Fernandez-Funez P, Golde TE. Short Aβ peptides attenuate Aβ42 toxicity in vivo. J Exp Med 2017; 215:283-301. [PMID: 29208777 PMCID: PMC5748850 DOI: 10.1084/jem.20170600] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/18/2017] [Accepted: 10/04/2017] [Indexed: 01/05/2023] Open
Abstract
Data demonstrate that short amyloid-β (Aβ) peptides are not toxic in vivo and can partially block toxicity associated with Aβ42 accumulation. Moore et al. further validate the use of γ-secretase modulators that lower Aβ42 and increase short Aβs as potential Alzheimer’s disease therapeutics. Processing of amyloid-β (Aβ) precursor protein (APP) by γ-secretase produces multiple species of Aβ: Aβ40, short Aβ peptides (Aβ37–39), and longer Aβ peptides (Aβ42–43). γ-Secretase modulators, a class of Alzheimer’s disease therapeutics, reduce production of the pathogenic Aβ42 but increase the relative abundance of short Aβ peptides. To evaluate the pathological relevance of these peptides, we expressed Aβ36–40 and Aβ42–43 in Drosophila melanogaster to evaluate inherent toxicity and potential modulatory effects on Aβ42 toxicity. In contrast to Aβ42, the short Aβ peptides were not toxic and, when coexpressed with Aβ42, were protective in a dose-dependent fashion. In parallel, we explored the effects of recombinant adeno-associated virus–mediated expression of Aβ38 and Aβ40 in mice. When expressed in nontransgenic mice at levels sufficient to drive Aβ42 deposition, Aβ38 and Aβ40 did not deposit or cause behavioral alterations. These studies indicate that treatments that lower Aβ42 by raising the levels of short Aβ peptides could attenuate the toxic effects of Aβ42.
Collapse
Affiliation(s)
- Brenda D Moore
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Jason Martin
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Lorena de Mena
- McKnight Brain Institute, University of Florida, Gainesville, FL.,Department of Neurology, University of Florida, Gainesville, FL
| | - Jonatan Sanchez
- McKnight Brain Institute, University of Florida, Gainesville, FL.,Department of Neurology, University of Florida, Gainesville, FL
| | - Pedro E Cruz
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Carolina Ceballos-Diaz
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Thomas B Ladd
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Yong Ran
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Yona Levites
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Thomas L Kukar
- Department of Pharmacology and Neurology, Emory University School of Medicine, Atlanta, GA
| | - Justin J Kurian
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL
| | - Edward H Koo
- Department of Neuroscience, University of California, San Diego, La Jolla, CA
| | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Christopher Janus
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL.,McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Diego Rincon-Limas
- McKnight Brain Institute, University of Florida, Gainesville, FL.,Department of Neurology, University of Florida, Gainesville, FL
| | - Pedro Fernandez-Funez
- Department of Biomedical Sciences, University of Minnesota School of Medicine, Duluth, MN
| | - Todd E Golde
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL .,McKnight Brain Institute, University of Florida, Gainesville, FL
| |
Collapse
|
47
|
Rittschof CC, Schirmeier S. Insect models of central nervous system energy metabolism and its links to behavior. Glia 2017; 66:1160-1175. [DOI: 10.1002/glia.23235] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/30/2017] [Accepted: 09/08/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Clare C. Rittschof
- Department of Entomology; College of Agriculture, Food, and the Environment, University of Kentucky; Lexington Kentucky
| | - Stefanie Schirmeier
- Institut für Neuro-und Verhaltensbiologie, University of Münster; Münster Germany
| |
Collapse
|
48
|
Richier B, Vijandi CDM, Mackensen S, Salecker I. Lapsyn controls branch extension and positioning of astrocyte-like glia in the Drosophila optic lobe. Nat Commun 2017; 8:317. [PMID: 28827667 PMCID: PMC5567088 DOI: 10.1038/s41467-017-00384-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 06/21/2017] [Indexed: 11/09/2022] Open
Abstract
Astrocytes have diverse, remarkably complex shapes in different brain regions. Their branches closely associate with neurons. Despite the importance of this heterogeneous glial cell type for brain development and function, the molecular cues controlling astrocyte branch morphogenesis and positioning during neural circuit assembly remain largely unknown. We found that in the Drosophila visual system, astrocyte-like medulla neuropil glia (mng) variants acquire stereotypic morphologies with columnar and layered branching patterns in a stepwise fashion from mid-metamorphosis onwards. Using knockdown and loss-of-function analyses, we uncovered a previously unrecognized role for the transmembrane leucine-rich repeat protein Lapsyn in regulating mng development. lapsyn is expressed in mng and cell-autonomously required for branch extension into the synaptic neuropil and anchoring of cell bodies at the neuropil border. Lapsyn works in concert with the fibroblast growth factor (FGF) pathway to promote branch morphogenesis, while correct positioning is essential for mng survival mediated by gliotrophic FGF signaling. How glial cells, such as astrocytes, acquire their characteristic morphology during development is poorly understood. Here the authors describe the morphogenesis of astrocyte-like glia in the Drosophila optic lobe, and through a RNAi screen, they identify a transmembrane LRR protein–Lapsyn–that plays a critical role in this process.
Collapse
Affiliation(s)
- Benjamin Richier
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.,The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | | | - Stefanie Mackensen
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.,University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, 48149, Muenster, Germany
| | - Iris Salecker
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
49
|
Mohylyak II, Chernyk YI. Functioning of glia and neurodegeneration in Drosophila melanogaster. CYTOL GENET+ 2017. [DOI: 10.3103/s0095452717030094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Tissue-specific transcription of the neuronal gene Lim3 affects Drosophila melanogaster lifespan and locomotion. Biogerontology 2017; 18:739-757. [DOI: 10.1007/s10522-017-9704-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/27/2017] [Indexed: 12/22/2022]
|