1
|
Jiménez-Martínez ML, Ramírez-Ahuja MDL, Saldaña-Torres DR, Martinez-Fierro ML, Delgado-Enciso I, Flores-Suarez AE, Reséndez-Pérez D, Guzmán-Velasco A, Rodríguez-Sánchez IP. De Novo miRNAs from Anisopteromalus calandrae (Hymenoptera: Pteromalidae) Conserved in the Order Hymenoptera. INSECTS 2024; 15:1007. [PMID: 39769609 PMCID: PMC11678685 DOI: 10.3390/insects15121007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
The parasitoid wasp Anisopteromalus calandrae (Howard) (Hymenoptera: Pteromalidae) has the potential for biological control against insect pests in stored grains, mainly of the orders Coleoptera and Lepidoptera. microRNAs (miRNAs) are small non-coding RNA fragments of importance in the regulation of gene translation in most physiological processes, and the study of miRNAs in wasps can be useful for understanding the physiology of these insects. The objective of this study was to evaluate for the first time the miRNomic profile of A. calandrae and to determine its conservation in five species of the order Hymenoptera (Apis mellifera, Dinoponera quadriceps, Nasonia giraulti, N. longicornis and N. vitripennis). Using molecular techniques and bioinformatics tools, a total of 108 miRNAs were identified (75 conserved between species and 34 de novo). These miRNAs were found to be related to embryogenesis, signaling, metabolic, biological and immune functions. The miRNomic signature of A. calandrae is important for the study of the physiology of wasps and the order Hymenoptera.
Collapse
Affiliation(s)
- Mariana Lizbeth Jiménez-Martínez
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 64460, Mexico; (M.L.J.-M.); (M.d.L.R.-A.); (D.R.S.-T.)
| | - María de Lourdes Ramírez-Ahuja
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 64460, Mexico; (M.L.J.-M.); (M.d.L.R.-A.); (D.R.S.-T.)
| | - Daniel Rafael Saldaña-Torres
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 64460, Mexico; (M.L.J.-M.); (M.d.L.R.-A.); (D.R.S.-T.)
| | | | - Ivan Delgado-Enciso
- Departamento de Medicina Molecular, Universidad de Colima, Escuela de Medicina, Colima 28040, Mexico;
| | - Adriana E. Flores-Suarez
- Laboratorio de Entomología Médica, Departamento de Zoología de Invertebrados, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 66455, Mexico;
| | - Diana Reséndez-Pérez
- Unidad de Biología del Desarrollo, Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas (Unidad C), Universidad Autónoma de Nuevo León, San Nicolás de Los Garza 66450, Mexico;
| | - Antonio Guzmán-Velasco
- Laboratorio de Conservación de Vida Silvestre y Desarrollo Sustentable, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 64460, Mexico
| | - Iram Pablo Rodríguez-Sánchez
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza 64460, Mexico; (M.L.J.-M.); (M.d.L.R.-A.); (D.R.S.-T.)
| |
Collapse
|
2
|
Wang T, Frank CA. Using Electrophysiology to Study Homeostatic Plasticity at the Drosophila Neuromuscular Junction. Cold Spring Harb Protoc 2024:pdb.top108393. [PMID: 38688539 PMCID: PMC11522024 DOI: 10.1101/pdb.top108393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The Drosophila melanogaster neuromuscular junction (NMJ) is a superb system for studying synapse function. Beyond that, the NMJ is also great for studying forms of synaptic plasticity. Over the last 25 years, Drosophila NMJ neuroscientists have pioneered understanding of a form of plasticity called homeostatic synaptic plasticity, which imparts functional stability on synaptic connections. The reason is straightforward: The NMJ has a robust capacity for stability. Moreover, many strategies that the NMJ uses to maintain appropriate levels of function are mirrored at other metazoan synapses. Here, we introduce core approaches that neurophysiologists use to study homeostatic synaptic plasticity at the peripheral Drosophila NMJ. We focus on methods to study a specific form of homeostatic plasticity termed presynaptic homeostatic potentiation (PHP), which is the most well-characterized one. Other forms such as presynaptic homeostatic depression and developmental forms of homeostasis are briefly discussed. Finally, we share lists of several dozen factors and conditions known to influence the execution of PHP.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C. 20007, USA
| | - C Andrew Frank
- Department of Anatomy and Cell Biology, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| |
Collapse
|
3
|
Bernard EIM, Towler BP, Rogoyski OM, Newbury SF. Characterisation of the in-vivo miRNA landscape in Drosophila ribonuclease mutants reveals Pacman-mediated regulation of the highly conserved let-7 cluster during apoptotic processes. Front Genet 2024; 15:1272689. [PMID: 38444757 PMCID: PMC10912645 DOI: 10.3389/fgene.2024.1272689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
The control of gene expression is a fundamental process essential for correct development and to maintain homeostasis. Many post-transcriptional mechanisms exist to maintain the correct levels of each RNA transcript within the cell. Controlled and targeted cytoplasmic RNA degradation is one such mechanism with the 5'-3' exoribonuclease Pacman (XRN1) and the 3'-5' exoribonuclease Dis3L2 playing crucial roles. Loss of function mutations in either Pacman or Dis3L2 have been demonstrated to result in distinct phenotypes, and both have been implicated in human disease. One mechanism by which gene expression is controlled is through the function of miRNAs which have been shown to be crucial for the control of almost all cellular processes. Although the biogenesis and mechanisms of action of miRNAs have been comprehensively studied, the mechanisms regulating their own turnover are not well understood. Here we characterise the miRNA landscape in a natural developing tissue, the Drosophila melanogaster wing imaginal disc, and assess the importance of Pacman and Dis3L2 on the abundance of miRNAs. We reveal a complex landscape of miRNA expression and show that whilst a null mutation in dis3L2 has a minimal effect on the miRNA expression profile, loss of Pacman has a profound effect with a third of all detected miRNAs demonstrating Pacman sensitivity. We also reveal a role for Pacman in regulating the highly conserved let-7 cluster (containing miR-100, let-7 and miR-125) and present a genetic model outlining a positive feedback loop regulated by Pacman which enhances our understanding of the apoptotic phenotype observed in Pacman mutants.
Collapse
Affiliation(s)
- Elisa I. M. Bernard
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Benjamin P. Towler
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
- School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Oliver M. Rogoyski
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Sarah F. Newbury
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
4
|
Mori M, Rosko A, Farnsworth J, Carrasco G, Broomandkhoshbacht P, Pareja-Navarro K, Pejmun Haghighi A. SimplyFire: An Open-Source, Customizable Software Application for the Analysis of Synaptic Events. eNeuro 2024; 11:ENEURO.0326-23.2023. [PMID: 38167616 PMCID: PMC10849045 DOI: 10.1523/eneuro.0326-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/30/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
We have developed an open-source software for neuroscientists to analyze electrophysiological recordings. Named SimplyFire, the software gives the users the flexibility to analyze a variety of recordings using an interactive graphical user interface or as an importable Python package. The software features a simple plugin structure that allows users to create and deploy various electrophysiology analysis tools. SimplyFire is pre-packaged with tools commonly used in electrophysiology, such as noise filtering, trace averaging, miniature analysis, and trace exporting. We discuss in detail the algorithm behind the different features of the analysis tool. We verify the accuracy of the algorithm by testing the software using computer-generated traces with known true values of the events. SimplyFire will be distributed under the GPLv3.0 license. The open nature of this software will allow interested investigators to modify and expand the software for additional capabilities as needed. We believe this software will not only compete with commercially available software packages but will also present a powerful tool to meet the current and unmet needs of electrophysiologists.
Collapse
Affiliation(s)
- Megumi Mori
- The Buck Institute for Research on Aging, Novato 94947, California
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco 94158, California
| | - Andrew Rosko
- The Buck Institute for Research on Aging, Novato 94947, California
| | - Jill Farnsworth
- The Buck Institute for Research on Aging, Novato 94947, California
| | | | | | | | | |
Collapse
|
5
|
Mallik B, Brusich DJ, Heyrman G, Frank CA. Precise mapping of one classic and three novel GluRIIA mutants in Drosophila melanogaster. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000784. [PMID: 37334199 PMCID: PMC10276266 DOI: 10.17912/micropub.biology.000784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/24/2023] [Accepted: 05/25/2023] [Indexed: 06/20/2023]
Abstract
Mutation of the Drosophila melanogaster GluRIIA gene or pharmacological agents targeting it are commonly used to assess homeostatic synaptic function at the larval neuromuscular junction (NMJ). The commonly used mutation, GluRIIA SP16 , is a null allele created by a large and imprecise excision of a P-element which affects GluRIIA and multiple upstream genes. Here we mapped the exact bounds of the GluRIIA SP16 allele, refined a multiplex PCR strategy for positive identification of GluRIIA SP16 in homozygous or heterozygous backgrounds, and sequenced and characterized three new CRISPR-generated GluRIIA mutants. We found the three new GluRIIA alleles are apparent nulls that lack GluRIIA immunofluorescence signal at the 3 rd instar larval NMJ and are predicted to cause premature truncations at the genetic level. Further, these new mutants have similar electrophysiological outcomes as GluRIIA SP16 , including reduced miniature excitatory postsynaptic potential (mEPSP) amplitude and frequency compared to controls, and they express robust homeostatic compensation as evidenced by normal excitatory postsynaptic potential (EPSP) amplitude and elevated quantal content. These findings and new tools extend the capacity of the D. melanogaster NMJ for assessment of synaptic function.
Collapse
Affiliation(s)
- Bhagaban Mallik
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States
| | - Douglas J Brusich
- Human Biology Department, University of Wisconsin–Green Bay, Green Bay, Wisconsin, United States
| | - Georgette Heyrman
- Human Biology Department, University of Wisconsin–Green Bay, Green Bay, Wisconsin, United States
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa, United States
- Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
6
|
Recent Advances and Future Potential of Long Non-Coding RNAs in Insects. Int J Mol Sci 2023; 24:ijms24032605. [PMID: 36768922 PMCID: PMC9917219 DOI: 10.3390/ijms24032605] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/31/2023] Open
Abstract
Over the last decade, long non-coding RNAs (lncRNAs) have witnessed a steep rise in interest amongst the scientific community. Because of their functional significance in several biological processes, i.e., alternative splicing, epigenetics, cell cycle, dosage compensation, and gene expression regulation, lncRNAs have transformed our understanding of RNA's regulatory potential. However, most knowledge concerning lncRNAs comes from mammals, and our understanding of the potential role of lncRNAs amongst insects remains unclear. Technological advances such as RNA-seq have enabled entomologists to profile several hundred lncRNAs in insect species, although few are functionally studied. This article will review experimentally validated lncRNAs from different insects and the lncRNAs identified via bioinformatic tools. Lastly, we will discuss the existing research challenges and the future of lncRNAs in insects.
Collapse
|
7
|
Liu J, Jin T, Ran L, Zhao Z, Zhu R, Xie G, Bi X. Profiling ATM regulated genes in Drosophila at physiological condition and after ionizing radiation. Hereditas 2022; 159:41. [PMID: 36271387 PMCID: PMC9587650 DOI: 10.1186/s41065-022-00254-9] [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: 06/06/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
Background ATM (ataxia-telangiectasia mutated) protein kinase is highly conserved in metazoan, and plays a critical role at DNA damage response, oxidative stress, metabolic stress, immunity, RNA biogenesis etc. Systemic profiling of ATM regulated genes, including protein-coding genes, miRNAs, and long non-coding RNAs, will greatly improve our understanding of ATM functions and its regulation. Results 1) differentially expressed protein-coding genes, miRNAs, and long non-coding RNAs in atm mutated flies were identified at physiological condition and after X-ray irradiation. 2) functions of differentially expressed genes in atm mutated flies, regardless of protein-coding genes or non-coding RNAs, are closely related with metabolic process, immune response, DNA damage response or oxidative stress. 3) these phenomena are persistent after irradiation. 4) there is a cross-talk regulation towards miRNAs by ATM, E2f1, and p53 during development and after irradiation. 5) knock-out flies or knock-down flies of most irradiation-induced miRNAs were sensitive to ionizing radiation. Conclusions We provide a valuable resource of protein-coding genes, miRNAs, and long non-coding RNAs, for understanding ATM functions and regulations. Our work provides the new evidence of inter-dependence among ATM-E2F1-p53 for the regulation of miRNAs. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-022-00254-9.
Collapse
Affiliation(s)
- Jun Liu
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Tianyu Jin
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Lanxi Ran
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Ze Zhao
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Rui Zhu
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Gangcai Xie
- School of Medicine, Nantong University, Nantong, 226001, China.
| | - Xiaolin Bi
- School of Medicine, Nantong University, Nantong, 226001, China. .,College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
8
|
Song C, Leahy SN, Rushton EM, Broadie K. RNA-binding FMRP and Staufen sequentially regulate the Coracle scaffold to control synaptic glutamate receptor and bouton development. Development 2022; 149:274991. [PMID: 35394012 PMCID: PMC9148565 DOI: 10.1242/dev.200045] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 03/23/2022] [Indexed: 12/16/2022]
Abstract
Both mRNA-binding Fragile X mental retardation protein (FMRP; Fmr1) and mRNA-binding Staufen regulate synaptic bouton formation and glutamate receptor (GluR) levels at the Drosophila neuromuscular junction (NMJ) glutamatergic synapse. Here, we tested whether these RNA-binding proteins act jointly in a common mechanism. We found that both dfmr1 and staufen mutants, and trans-heterozygous double mutants, displayed increased synaptic bouton formation and GluRIIA accumulation. With cell-targeted RNA interference, we showed a downstream Staufen role within postsynaptic muscle. With immunoprecipitation, we showed that FMRP binds staufen mRNA to stabilize postsynaptic transcripts. Staufen is known to target actin-binding, GluRIIA anchor Coracle, and we confirmed that Staufen binds to coracle mRNA. We found that FMRP and Staufen act sequentially to co-regulate postsynaptic Coracle expression, and showed that Coracle, in turn, controls GluRIIA levels and synaptic bouton development. Consistently, we found that dfmr1, staufen and coracle mutants elevate neurotransmission strength. We also identified that FMRP, Staufen and Coracle all suppress pMad activation, providing a trans-synaptic signaling linkage between postsynaptic GluRIIA levels and presynaptic bouton development. This work supports an FMRP-Staufen-Coracle-GluRIIA-pMad pathway regulating structural and functional synapse development.
Collapse
Affiliation(s)
- Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Emma M. Rushton
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA,Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN 37235, USA,Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA,Author for correspondence ()
| |
Collapse
|
9
|
An Emerging Role for Epigenetics in Cerebral Palsy. J Pers Med 2021; 11:jpm11111187. [PMID: 34834539 PMCID: PMC8625874 DOI: 10.3390/jpm11111187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 12/29/2022] Open
Abstract
Cerebral palsy is a set of common, severe, motor disabilities categorized by a static, nondegenerative encephalopathy arising in the developing brain and associated with deficits in movement, posture, and activity. Spastic CP, which is the most common type, involves high muscle tone and is associated with altered muscle function including poor muscle growth and contracture, increased extracellular matrix deposition, microanatomic disruption, musculoskeletal deformities, weakness, and difficult movement control. These muscle-related manifestations of CP are major causes of progressive debilitation and frequently require intensive surgical and therapeutic intervention to control. Current clinical approaches involve sophisticated consideration of biomechanics, radiologic assessments, and movement analyses, but outcomes remain difficult to predict. There is a need for more precise and personalized approaches involving omics technologies, data science, and advanced analytics. An improved understanding of muscle involvement in spastic CP is needed. Unfortunately, the fundamental mechanisms and molecular pathways contributing to altered muscle function in spastic CP are only partially understood. In this review, we outline evidence supporting the emerging hypothesis that epigenetic phenomena play significant roles in musculoskeletal manifestations of CP.
Collapse
|
10
|
Woods BJ, Van Vactor D. miRNA: local guardians of presynaptic function in plasticity and disease. RNA Biol 2021; 18:1014-1024. [PMID: 33586621 PMCID: PMC8216186 DOI: 10.1080/15476286.2020.1871214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 12/27/2022] Open
Abstract
Environmental fitness is an essential component of animal survival. Fitness is achieved through responsive physiological plasticity of tissues across the entire body, and particularly in the nervous system. At the molecular level, neural plasticity is mediated via gene-environmental interactions whereby developmental cues and experience dependent input adapt neuronal function to ever changing demands. To this end, neuronal gene regulation must be coupled to changes in neural activity. Seminal discoveries of the 20th century demonstrated neural activity modifies gene expression through calcium-dependent gene transcription. Building on this model, recent work over the last two decades shows that mRNA products of transcriptional programming continue to be regulated in the neuron through the activity-dependent post-transcriptional action of microRNAs (miRNAs). miRNAs are special post-transcriptional regulators that can tune gene expression within the spatial and temporal requirements of synaptic compartments. This mode of gene regulation has proven to be essential for synaptic function and plasticity as miRNA loss of function is highly associated with neural disease. In this review we will discuss current perspective on the link between presynaptic plasticity and miRNA biogenesis in the neuron.
Collapse
Affiliation(s)
- Brandon J. Woods
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - David Van Vactor
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Changes in Presynaptic Gene Expression during Homeostatic Compensation at a Central Synapse. J Neurosci 2021; 41:3054-3067. [PMID: 33608385 PMCID: PMC8026347 DOI: 10.1523/jneurosci.2979-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/02/2022] Open
Abstract
Homeostatic matching of pre- and postsynaptic function has been observed in many species and neural structures, but whether transcriptional changes contribute to this form of trans-synaptic coordination remains unknown. To identify genes whose expression is altered in presynaptic neurons as a result of perturbing postsynaptic excitability, we applied a transcriptomics-friendly, temperature-inducible Kir2.1-based activity clamp at the first synaptic relay of the Drosophila olfactory system, a central synapse known to exhibit trans-synaptic homeostatic matching. Twelve hours after adult-onset suppression of activity in postsynaptic antennal lobe projection neurons of males and females, we detected changes in the expression of many genes in the third antennal segment, which houses the somata of presynaptic olfactory receptor neurons. These changes affected genes with roles in synaptic vesicle release and synaptic remodeling, including several implicated in homeostatic plasticity at the neuromuscular junction. At 48 h and beyond, the transcriptional landscape tilted toward protein synthesis, folding, and degradation; energy metabolism; and cellular stress defenses, indicating that the system had been pushed to its homeostatic limits. Our analysis suggests that similar homeostatic machinery operates at peripheral and central synapses and identifies many of its components. The presynaptic transcriptional response to genetically targeted postsynaptic perturbations could be exploited for the construction of novel connectivity tracing tools. SIGNIFICANCE STATEMENT Homeostatic feedback mechanisms adjust intrinsic and synaptic properties of neurons to keep their average activity levels constant. We show that, at a central synapse in the fruit fly brain, these mechanisms include changes in presynaptic gene expression that are instructed by an abrupt loss of postsynaptic excitability. The trans-synaptically regulated genes have roles in synaptic vesicle release and synapse remodeling; protein synthesis, folding, and degradation; and energy metabolism. Our study establishes a role for transcriptional changes in homeostatic synaptic plasticity, points to mechanistic commonalities between peripheral and central synapses, and potentially opens new opportunities for the development of connectivity-based gene expression systems.
Collapse
|
12
|
miR-92a Suppresses Mushroom Body-Dependent Memory Consolidation in Drosophila. eNeuro 2020; 7:ENEURO.0224-20.2020. [PMID: 32737186 PMCID: PMC7642123 DOI: 10.1523/eneuro.0224-20.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) fine tune gene expression to regulate many aspects of nervous system physiology. Here, we show that miR-92a suppresses memory consolidation that occurs in the αβ and γ mushroom body neurons (MBns) of Drosophila, making miR-92a a memory suppressor miRNA. Bioinformatics analyses suggested that mRNAs encoding kinesin heavy chain 73 (KHC73), a protein that belongs to Kinesin-3 family of anterograde motor proteins, may be a functional target of miR-92a. Behavioral studies that employed expression of khc73 with and without its 3' untranslated region (UTR) containing miR-92a target sites, luciferase assays in HEK cells with reporters containing wild-type and mutant target sequences in the khc73 3'UTR, and immunohistochemistry experiments involving KHC73 expression with and without the wild-type khc73 3'UTR, all point to the conclusion that khc73 is a major target of miR-92a in its functional role as a miRNA memory suppressor gene.
Collapse
|
13
|
Zhang X, Lyu J, Jin X, Yang W, Lou Z, Xi Y, Yang X, Ge W. A motor neuron protective role of miR-969 mediated by the transcription factor kay. RNA Biol 2020; 17:1277-1283. [PMID: 32397794 DOI: 10.1080/15476286.2020.1757897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Maintenance of motor neuron structure and function is crucial in development and motor behaviour. However, the genetic regulatory mechanism of motor neuron function remains less well understood. In the present study, we identify a novel neuroprotective role of the microRNA miR-969 in Drosophila motor neurons. miR-969 is highly expressed in motor neurons. Loss of miR-969 results in early-onset and age-progressive locomotion impairment. Flies lacking miR-969 also exhibit shortened lifespan. Moreover, miR-969 is required in motor neurons. We further identify kay as a functionally important target of miR-969. Together, our results indicate that miR-969 can protect motor neuron function by limiting kay activity in Drosophila.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Jialan Lyu
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Xiaoye Jin
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Weiwei Yang
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Ziwei Lou
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Yongmei Xi
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Xiaohang Yang
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| | - Wanzhong Ge
- Division of Human Reproduction and Developmental Genetics, Women's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China.,Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine , Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
McNeill EM, Warinner C, Alkins S, Taylor A, Heggeness H, DeLuca TF, Fulga TA, Wall DP, Griffith LC, Van Vactor D. The conserved microRNA miR-34 regulates synaptogenesis via coordination of distinct mechanisms in presynaptic and postsynaptic cells. Nat Commun 2020; 11:1092. [PMID: 32107390 PMCID: PMC7046720 DOI: 10.1038/s41467-020-14761-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/31/2020] [Indexed: 01/30/2023] Open
Abstract
Micro(mi)RNA-based post-transcriptional regulatory mechanisms have been broadly implicated in the assembly and modulation of synaptic connections required to shape neural circuits, however, relatively few specific miRNAs have been identified that control synapse formation. Using a conditional transgenic toolkit for competitive inhibition of miRNA function in Drosophila, we performed an unbiased screen for novel regulators of synapse morphogenesis at the larval neuromuscular junction (NMJ). From a set of ten new validated regulators of NMJ growth, we discovered that miR-34 mutants display synaptic phenotypes and cell type-specific functions suggesting distinct downstream mechanisms in the presynaptic and postsynaptic compartments. A search for conserved downstream targets for miR-34 identified the junctional receptor CNTNAP4/Neurexin-IV (Nrx-IV) and the membrane cytoskeletal effector Adducin/Hu-li tai shao (Hts) as proteins whose synaptic expression is restricted by miR-34. Manipulation of miR-34, Nrx-IV or Hts-M function in motor neurons or muscle supports a model where presynaptic miR-34 inhibits Nrx-IV to influence active zone formation, whereas, postsynaptic miR-34 inhibits Hts to regulate the initiation of bouton formation from presynaptic terminals. Although micro(mi)RNA-based post-transcriptional regulatory mechanisms have been implicated in the assembly and modulation of synaptic connections, few miRNAs have been identified that control synapse formation. Here, authors performed an unbiased screen for novel regulators of synapse morphogenesis at the Drosophila larval neuromuscular junction and discovered that miR-34 inhibits Nrx-IV to influence active zone formation, whereas, postsynaptic miR-34 inhibits Hts to regulate the initiation of bouton formation from presynaptic terminals.
Collapse
Affiliation(s)
- Elizabeth M McNeill
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.,Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Chloe Warinner
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephen Alkins
- Department of Biology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, 02454, USA
| | - Alicia Taylor
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.,Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Hansine Heggeness
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| | - Todd F DeLuca
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| | - Tudor A Fulga
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.,Weatherall Institute, Oxford University, Oxford, UK
| | - Dennis P Wall
- Department of Pediatrics, Division of Systems Medicine, Stanford University, Palo Alto, CA, 94305, USA
| | - Leslie C Griffith
- Department of Biology and Volen National Center for Complex Systems, Brandeis University, Waltham, MA, 02454, USA
| | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
15
|
Abstract
Locomotion is an ancient and fundamental output of the nervous system required for animals to perform many other complex behaviors. Although the formation of motor circuits is known to be under developmental control of transcriptional mechanisms that define the fates and connectivity of the many neurons, glia and muscle constituents of these circuits, relatively little is known about the role of post-transcriptional regulation of locomotor behavior. MicroRNAs have emerged as a potentially rich source of modulators for neural development and function. In order to define the microRNAs required for normal locomotion in Drosophila melanogaster, we utilized a set of transgenic Gal4-dependent competitive inhibitors (microRNA sponges, or miR-SPs) to functionally assess ca. 140 high-confidence Drosophila microRNAs using automated quantitative movement tracking systems followed by multiparametric analysis. Using ubiquitous expression of miR-SP constructs, we identified a large number of microRNAs that modulate aspects of normal baseline adult locomotion. Addition of temperature-dependent Gal80 to identify microRNAs that act during adulthood revealed that the majority of these microRNAs play developmental roles. Comparison of ubiquitous and neural-specific miR-SP expression suggests that most of these microRNAs function within the nervous system. Parallel analyses of spontaneous locomotion in adults and in larvae also reveal that very few of the microRNAs required in the adult overlap with those that control the behavior of larval motor circuits. These screens suggest that a rich regulatory landscape underlies the formation and function of motor circuits and that many of these mechanisms are stage and/or parameter-specific.
Collapse
|
16
|
Dubes S, Favereaux A, Thoumine O, Letellier M. miRNA-Dependent Control of Homeostatic Plasticity in Neurons. Front Cell Neurosci 2019; 13:536. [PMID: 31866828 PMCID: PMC6906196 DOI: 10.3389/fncel.2019.00536] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/19/2019] [Indexed: 11/13/2022] Open
Abstract
Homeostatic plasticity is a form of plasticity in which neurons compensate for changes in neuronal activity through the control of key physiological parameters such as the number and the strength of their synaptic inputs and intrinsic excitability. Recent studies revealed that miRNAs, which are small non-coding RNAs repressing mRNA translation, participate in this process by controlling the translation of multiple effectors such as glutamate transporters, receptors, signaling molecules and voltage-gated ion channels. In this review, we present and discuss the role of miRNAs in both cell-wide and compartmentalized forms of homeostatic plasticity as well as their implication in pathological processes associated with homeostatic failure.
Collapse
Affiliation(s)
- Sandra Dubes
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Alexandre Favereaux
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Olivier Thoumine
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Mathieu Letellier
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| |
Collapse
|
17
|
Signal Exchange through Extracellular Vesicles in Neuromuscular Junction Establishment and Maintenance: From Physiology to Pathology. Int J Mol Sci 2019; 20:ijms20112804. [PMID: 31181747 PMCID: PMC6600513 DOI: 10.3390/ijms20112804] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
Neuromuscular junction (NMJ) formation involves morphological changes both in motor terminals and muscle membrane. The molecular mechanisms leading to NMJ formation and maintenance have not yet been fully elucidated. During the last decade, it has become clear that virtually all cells release different types of extracellular vesicles (EVs), which can be taken up by nearby or distant cells modulating their activity. Initially, EVs were associated to a mechanism involved in the elimination of unwanted material; subsequent evidence demonstrated that exosomes, and more in general EVs, play a key role in intercellular communication by transferring proteins, lipids, DNA and RNA to target cells. Recently, EVs have emerged as potent carriers for Wnt, bone morphogenetic protein, miRNA secretion and extracellular traveling. Convincing evidence demonstrates that presynaptic terminals release exosomes that are taken up by muscle cells, and these exosomes can modulate synaptic plasticity in the recipient muscle cell in vivo. Furthermore, recent data highlighted that EVs could also be a potential cause of neurodegenerative disorders. Indeed, mutant SOD1, TDP-43 and FUS/TLS can be secreted by neural cells packaged into EVs and enter in neighboring neural cells, contributing to the onset and severity of the disease.
Collapse
|
18
|
Exposure to enriched environment rescues anxiety-like behavior and miRNA deregulated expression induced by perinatal malnutrition while altering oligodendrocyte morphology. Neuroscience 2019; 408:115-134. [PMID: 30904666 DOI: 10.1016/j.neuroscience.2019.03.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/21/2022]
Abstract
Maternal malnutrition is one of the major early-life adversities affecting the development of newborn's brain and is associated with an increased risk to acquire cognitive and emotional deficiencies later in life. Studies in rodents have demonstrated that exposure to an enriched environment (EE) can reverse the negative consequences of early adversities. However, rescue of emotional disorders caused by perinatal malnutrition and the mechanisms involved has not been determined. We hypothesized that exposure to an EE may attenuate the anxiety-like disorders observed in mice subjected to perinatal protein malnutrition and that this could be mediated by epigenetic mechanisms. Male CF-1 mice were subject to perinatal protein malnutrition until weaning and then exposed to an EE for 5 weeks after which small RNA-seq was performed. In parallel, dark-light box and elevated plus maze tests were conducted to evaluate anxiety traits. We found that exposure to an EE reverses the anxiety-like behavior in malnourished mice. This reversal is paralleled by the expression of three miRNAs that become dysregulated by perinatal malnutrition (miR-187-3p, miR-369-3p and miR-132-3p). The predicted mRNA targets of these miRNAs are mostly related to axon guidance pathway. Accordingly, we also found that perinatal malnutrition leads to reduction in the cingulum size and altered oligodendrocyte morphology. These results suggest that EE-rescue of anxiety disorders derived from perinatal malnutrition is mediated by the modulation of miRNAs associated with the regulation of genes involved in axonal guidance.
Collapse
|
19
|
Böhme MA, McCarthy AW, Grasskamp AT, Beuschel CB, Goel P, Jusyte M, Laber D, Huang S, Rey U, Petzoldt AG, Lehmann M, Göttfert F, Haghighi P, Hell SW, Owald D, Dickman D, Sigrist SJ, Walter AM. Rapid active zone remodeling consolidates presynaptic potentiation. Nat Commun 2019; 10:1085. [PMID: 30842428 PMCID: PMC6403334 DOI: 10.1038/s41467-019-08977-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 02/07/2019] [Indexed: 01/22/2023] Open
Abstract
Neuronal communication across synapses relies on neurotransmitter release from presynaptic active zones (AZs) followed by postsynaptic transmitter detection. Synaptic plasticity homeostatically maintains functionality during perturbations and enables memory formation. Postsynaptic plasticity targets neurotransmitter receptors, but presynaptic mechanisms regulating the neurotransmitter release apparatus remain largely enigmatic. By studying Drosophila neuromuscular junctions (NMJs) we show that AZs consist of nano-modular release sites and identify a molecular sequence that adds modules within minutes of inducing homeostatic plasticity. This requires cognate transport machinery and specific AZ-scaffolding proteins. Structural remodeling is not required for immediate potentiation of neurotransmitter release, but necessary to sustain potentiation over longer timescales. Finally, mutations in Unc13 disrupting homeostatic plasticity at the NMJ also impair short-term memory when central neurons are targeted, suggesting that both plasticity mechanisms utilize Unc13. Together, while immediate synaptic potentiation capitalizes on available material, it triggers the coincident incorporation of modular release sites to consolidate synaptic potentiation.
Collapse
Affiliation(s)
- Mathias A Böhme
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin, 10117, Berlin, Germany.,Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Anthony W McCarthy
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Andreas T Grasskamp
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité Universitätsmedizin, 10117, Berlin, Germany
| | - Christine B Beuschel
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, 10117, Berlin, Germany.,Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Meida Jusyte
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Desiree Laber
- Institut für Neurophysiologie, Charité Universitätsmedizin, 10117, Berlin, Germany
| | - Sheng Huang
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Ulises Rey
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany.,Department of Theory and Bio-systems, Max Planck Institute of Colloids and Interfaces, Science Park Golm, 14424, Potsdam, Germany
| | - Astrid G Petzoldt
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Fabian Göttfert
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | | | - Stefan W Hell
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - David Owald
- Institut für Neurophysiologie, Charité Universitätsmedizin, 10117, Berlin, Germany
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Stephan J Sigrist
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, 10117, Berlin, Germany. .,Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany.
| | - Alexander M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany.
| |
Collapse
|
20
|
Seong KM, Coates BS, Pittendrigh BR. Impacts of Sub-lethal DDT Exposures on microRNA and Putative Target Transcript Expression in DDT Resistant and Susceptible Drosophila melanogaster Strains. Front Genet 2019; 10:45. [PMID: 30804985 PMCID: PMC6370691 DOI: 10.3389/fgene.2019.00045] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/21/2019] [Indexed: 01/06/2023] Open
Abstract
Ten constitutively differentially expressed miRNAs were previously described between DDT-resistant 91-R and -susceptible control Drosophila melanogaster strains, and among their predicted target genes were those associated with metabolic DDT resistance mechanisms. The present study evaluated the inducibility of miRNA expression and putative downstream regulation of cytochrome P450s in response to DDT exposure in a time-dependent manner in 91-R and the susceptible Canton-S strain. Specifically, RT-qPCR analysis showed that DDT exposures led to the significant down-regulation (repression) of miR-310-3p, miR-311-3p, miR-312-3p, miR-313-3p, and miR-92a-3p levels in Canton-S. This is contrasted with the lack of significant changes in 91-R at most time-points following DDT exposure. The levels of expression among miRNAs exhibited opposite expression patterns compared to their corresponding putative target cytochrome P450s at the same time points after DDT exposure. Collectively, results from this study suggest that miR-310-3p, miR-311-3p, miR-312-3p, miR-313-3p, and miR-92a-3p might have a potential role in the control of DDT detoxification through the post-transcriptional regulation of target cytochrome P450s in Canton-S. Conversely, the lack of significant changes of these same miRNAs in 91-R following DDT-exposure suggests a possible adaptive mutation that removes repressive control mechanisms. These data are important for the understanding impact of adaptive changes in miRNA expression on post-transcriptional regulatory mechanism involved in the evolution of DDT resistance in 91-R.
Collapse
Affiliation(s)
- Keon Mook Seong
- Department of Entomology, Michigan State University, East Lansing, MI, United States
| | - Brad S Coates
- USDA-ARS, Corn Insects and Crop Genetics Research Unit, Ames, IA, United States
| | - Barry R Pittendrigh
- Department of Entomology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
21
|
Kaschula R, Pinho S, Alonso CR. MicroRNA-dependent regulation of Hox gene expression sculpts fine-grain morphological patterns in a Drosophila appendage. Development 2018; 145:dev.161133. [PMID: 30143542 PMCID: PMC6215401 DOI: 10.1242/dev.161133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 08/06/2018] [Indexed: 11/26/2022]
Abstract
Disruptions of normal Hox gene expression can lead to severe morphological defects, revealing a link between the regulation of Hox expression and pattern formation. Here, we explore these links, focusing on the impact of microRNA regulation on the expression of the Drosophila Hox gene Ultrabithorax (Ubx) during haltere development. Through a combination of bioinformatic and transcriptomic analyses, we identify the miR-310/313 cluster (miR-310C) as a candidate regulator of Ubx. Several experiments confirm this. First, miR-310C and Ubx protein show complementary expression patterns in haltere imaginal discs; second, artificial activation of miR-310C expression in haltere discs leads to Ubx-like phenotypes. Third, expression of a fluorescent reporter bearing Ubx 3′UTR sequences is reduced when co-expressed with miR-310C. Fourth, deletion of miR-310C leads to Ubx upregulation and changes the array of mechanosensory sensilla at the base of the haltere. Fifth, an artificial increase of Ubx levels within the miR-310C expression domain phenocopies the mechanosensory defects observed in miR-310C mutants. We propose that miR-310C-mediated repression delimits Ubx fine-grain expression, contributing to the sculpting of complex morphologies in the Drosophila haltere with implications for flight control. Our work reveals a novel role of microRNA regulation in the control of Hox gene expression with impact on morphology. Summary: A novel role of microRNAs in the control of Hox gene expression with impact on the development of morphology in Drosophila, suggesting that microRNAs might play similar roles in other bilaterians.
Collapse
Affiliation(s)
- Richard Kaschula
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Sofia Pinho
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Claudio R Alonso
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| |
Collapse
|
22
|
Harris KP, Littleton JT, Stewart BA. Postsynaptic Syntaxin 4 negatively regulates the efficiency of neurotransmitter release. J Neurogenet 2018; 32:221-229. [PMID: 30175640 PMCID: PMC6317344 DOI: 10.1080/01677063.2018.1501372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022]
Abstract
Signaling from the postsynaptic compartment regulates multiple aspects of synaptic development and function. Syntaxin 4 (Syx4) is a plasma membrane t-SNARE that promotes the growth and plasticity of Drosophila neuromuscular junctions (NMJs) by regulating the localization of key synaptic proteins in the postsynaptic compartment. Here, we describe electrophysiological analyses and report that loss of Syx4 leads to enhanced neurotransmitter release, despite a decrease in the number of active zones. We describe a requirement for postsynaptic Syx4 in regulating several presynaptic parameters, including Ca2+ cooperativity and the abundance of the presynaptic calcium channel Cacophony (Cac) at active zones. These findings indicate Syx4 negatively regulates presynaptic neurotransmitter release through a retrograde signaling mechanism from the postsynaptic compartment.
Collapse
Affiliation(s)
- Kathryn P Harris
- a Department of Biology , University of Toronto Mississauga , Mississauga , ON, Canada
- b Department of Cell and Systems Biology , University of Toronto , Toronto , ON, Canada
| | - J Troy Littleton
- c The Picower Institute for Learning and Memory , Massachusetts Institute of Technology , Cambridge , MA , USA
- d Department of Biology , Massachusetts Institute of Technology , Cambridge , MA , USA
- e Department of Brain and Cognitive Sciences , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Bryan A Stewart
- a Department of Biology , University of Toronto Mississauga , Mississauga , ON, Canada
- b Department of Cell and Systems Biology , University of Toronto , Toronto , ON, Canada
| |
Collapse
|
23
|
Goel P, Li X, Dickman D. Disparate Postsynaptic Induction Mechanisms Ultimately Converge to Drive the Retrograde Enhancement of Presynaptic Efficacy. Cell Rep 2018; 21:2339-2347. [PMID: 29186673 DOI: 10.1016/j.celrep.2017.10.116] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/13/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022] Open
Abstract
Retrograde signaling systems are fundamental modes of communication synapses utilize to dynamically and adaptively modulate activity. However, the inductive mechanisms that gate retrograde communication in the postsynaptic compartment remain enigmatic. We have investigated retrograde signaling at the Drosophila neuromuscular junction, where three seemingly disparate perturbations to the postsynaptic cell trigger a similar enhancement in presynaptic neurotransmitter release. We show that the same presynaptic genetic machinery and enhancements in active zone structure are utilized by each inductive pathway. However, all three induction mechanisms differ in temporal, translational, and CamKII activity requirements to initiate retrograde signaling in the postsynaptic cell. Intriguingly, pharmacological blockade of postsynaptic glutamate receptors, and not calcium influx through these receptors, is necessary and sufficient to induce rapid retrograde homeostatic signaling through CamKII. Thus, three distinct induction mechanisms converge on the same retrograde signaling system to drive the homeostatic strengthening of presynaptic neurotransmitter release.
Collapse
Affiliation(s)
- Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA; Graduate Program in Molecular Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
24
|
Differentially expressed microRNAs associated with changes of transcript levels in detoxification pathways and DDT-resistance in the Drosophila melanogaster strain 91-R. PLoS One 2018; 13:e0196518. [PMID: 29698530 PMCID: PMC5919617 DOI: 10.1371/journal.pone.0196518] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
Dichloro-diphenyl-trichloroethane (DDT) resistance among arthropod species is a model for understanding the molecular adaptations in response to insecticide exposures. Previous studies reported that DDT resistance may involve one or multiple detoxification genes, such as cytochrome P450 monooxygenases (P450s), glutathione S-transferases (GSTs), esterases, and ATP binding cassette (ABC) transporters, or changes in the voltage-sensitive sodium channel. However, the possible involvement of microRNAs (miRNAs) in the post-transcriptional regulation of genes associated with DDT resistance in the Drosophila melanogaster strain 91-R remains poorly understood. In this study, the majority of the resulting miRNAs discovered in small RNA libraries from 91-R and the susceptible control strain, 91-C, ranged from 16-25 nt, and contained 163 precursors and 256 mature forms of previously-known miRNAs along with 17 putative novel miRNAs. Quantitative analyses predicted the differential expression of ten miRNAs between 91-R and 91-C, and, based on Gene Ontology and pathway analysis, these ten miRNAs putatively target transcripts encoding proteins involved in detoxification mechanisms. RT-qPCR validated an inverse correlation between levels of differentially-expressed miRNAs and their putatively targeted transcripts, which implies a role of these miRNAs in the differential regulation of detoxification pathways in 91-R compared to 91-C. This study provides evidence associating the differential expression of miRNAs in response to multigenerational DDT selection in Drosophila melanogaster and provides important clues for understanding the possible roles of miRNAs in mediating insecticide resistance traits.
Collapse
|
25
|
Li X, Goel P, Chen C, Angajala V, Chen X, Dickman DK. Synapse-specific and compartmentalized expression of presynaptic homeostatic potentiation. eLife 2018; 7:34338. [PMID: 29620520 PMCID: PMC5927770 DOI: 10.7554/elife.34338] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/04/2018] [Indexed: 01/23/2023] Open
Abstract
Postsynaptic compartments can be specifically modulated during various forms of synaptic plasticity, but it is unclear whether this precision is shared at presynaptic terminals. Presynaptic homeostatic plasticity (PHP) stabilizes neurotransmission at the Drosophila neuromuscular junction, where a retrograde enhancement of presynaptic neurotransmitter release compensates for diminished postsynaptic receptor functionality. To test the specificity of PHP induction and expression, we have developed a genetic manipulation to reduce postsynaptic receptor expression at one of the two muscles innervated by a single motor neuron. We find that PHP can be induced and expressed at a subset of synapses, over both acute and chronic time scales, without influencing transmission at adjacent release sites. Further, homeostatic modulations to CaMKII, vesicle pools, and functional release sites are compartmentalized and do not spread to neighboring pre- or post-synaptic structures. Thus, both PHP induction and expression mechanisms are locally transmitted and restricted to specific synaptic compartments. Everything we think and do is the result of communication between neurons. This communication takes place at junctions called synapses. When two nerve cells or neurons communicate at a synapse, the output terminal of the first cell releases a chemical called a neurotransmitter. This binds to receiver proteins, or receptors, on the second cell. When this communication is interrupted, synapses can adapt to maintain a stable dialogue between them. This can occur in two ways. Either the first neuron starts to release more neurotransmitter from its output terminal, or the second neuron produces extra receptors with which to detect the neurotransmitter. But how specific are these changes? The brain contains far more synapses than neurons because each neuron can form synapses with many other cells. Can a neuron adjust how much of the neurotransmitter it releases at some of its synapses while leaving the others unchanged? Li et al. have now addressed this question by studying a special type of synapse that forms between neurons and muscles, known as a neuromuscular junction. At one particular neuromuscular junction in fruit flies, a single neuron splits into two output terminals, each of which forms a synapse with a different muscle. Li et al. show that when the number of neurotransmitter receptors in one of the muscles is artificially reduced, the associated output terminal compensates by increasing its neurotransmitter release. By contrast, the other output terminal remains unaffected. This suggests that a neuron can induce remarkably specific changes in a subset of its synapses. This discovery paves the way towards identifying the smallest possible unit of change that can occur in the neurons’ ability to communicate. This unit may in turn be the smallest change that can support learning. Such knowledge will help us understand how the nervous system processes and stabilizes information transfer, both in health and after injury or disease.
Collapse
Affiliation(s)
- Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, United States.,Neuroscience Graduate Program, University of Southern California, California, United States
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, United States.,Graduate Program in Molecular and Computational Biology, University of Southern California, California, United States
| | - Catherine Chen
- Department of Neurobiology, University of Southern California, Los Angeles, United States
| | | | - Xun Chen
- Neuroscience Graduate Program, University of Southern California, California, United States
| | | |
Collapse
|
26
|
Liao EH, Gray L, Tsurudome K, El-Mounzer W, Elazzouzi F, Baim C, Farzin S, Calderon MR, Kauwe G, Haghighi AP. Kinesin Khc-73/KIF13B modulates retrograde BMP signaling by influencing endosomal dynamics at the Drosophila neuromuscular junction. PLoS Genet 2018; 14:e1007184. [PMID: 29373576 PMCID: PMC5802963 DOI: 10.1371/journal.pgen.1007184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 02/07/2018] [Accepted: 01/03/2018] [Indexed: 11/18/2022] Open
Abstract
Retrograde signaling is essential for neuronal growth, function and survival; however, we know little about how signaling endosomes might be directed from synaptic terminals onto retrograde axonal pathways. We have identified Khc-73, a plus-end directed microtubule motor protein, as a regulator of sorting of endosomes in Drosophila larval motor neurons. The number of synaptic boutons and the amount of neurotransmitter release at the Khc-73 mutant larval neuromuscular junction (NMJ) are normal, but we find a significant decrease in the number of presynaptic release sites. This defect in Khc-73 mutant larvae can be genetically enhanced by a partial genetic loss of Bone Morphogenic Protein (BMP) signaling or suppressed by activation of BMP signaling in motoneurons. Consistently, activation of BMP signaling that normally enhances the accumulation of phosphorylated form of BMP transcription factor Mad in the nuclei, can be suppressed by genetic removal of Khc-73. Using a number of assays including live imaging in larval motor neurons, we show that loss of Khc-73 curbs the ability of retrograde-bound endosomes to leave the synaptic area and join the retrograde axonal pathway. Our findings identify Khc-73 as a regulator of endosomal traffic at the synapse and modulator of retrograde BMP signaling in motoneurons.
Collapse
Affiliation(s)
- Edward H. Liao
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Lindsay Gray
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Kazuya Tsurudome
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | | | - Fatima Elazzouzi
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Christopher Baim
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Sarah Farzin
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Mario R. Calderon
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - A. Pejmun Haghighi
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Department of Physiology, McGill University, Montreal, QC, Canada
- * E-mail:
| |
Collapse
|
27
|
Dey S, Banker G, Ray K. Anterograde Transport of Rab4-Associated Vesicles Regulates Synapse Organization in Drosophila. Cell Rep 2017; 18:2452-2463. [PMID: 28273459 DOI: 10.1016/j.celrep.2017.02.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 12/19/2016] [Accepted: 02/09/2017] [Indexed: 11/29/2022] Open
Abstract
Local endosomal recycling at synapses is essential to maintain neurotransmission. Rab4GTPase, found on sorting endosomes, is proposed to balance the flow of vesicles among endocytic, recycling, and degradative pathways in the presynaptic compartment. Here, we report that Rab4-associated vesicles move bidirectionally in Drosophila axons but with an anterograde bias, resulting in their moderate enrichment at the synaptic region of the larval ventral ganglion. Results from FK506 binding protein (FKBP) and FKBP-Rapamycin binding domain (FRB) conjugation assays in rat embryonic fibroblasts together with genetic analyses in Drosophila indicate that an association with Kinesin-2 (mediated by the tail domain of Kinesin-2α/KIF3A/KLP64D subunit) moves Rab4-associated vesicles toward the synapse. Reduction in the anterograde traffic of Rab4 causes an expansion of the volume of the synapse-bearing region in the ventral ganglion and increases the motility of Drosophila larvae. These results suggest that Rab4-dependent vesicular traffic toward the synapse plays a vital role in maintaining synaptic balance in this neuronal network.
Collapse
Affiliation(s)
- Swagata Dey
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| | - Gary Banker
- Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR 97239, USA
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India.
| |
Collapse
|
28
|
Liufu Z, Zhao Y, Guo L, Miao G, Xiao J, Lyu Y, Chen Y, Shi S, Tang T, Wu CI. Redundant and incoherent regulations of multiple phenotypes suggest microRNAs' role in stability control. Genome Res 2017; 27:1665-1673. [PMID: 28904014 PMCID: PMC5630030 DOI: 10.1101/gr.222505.117] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022]
Abstract
Each microRNA (miRNA) represses a web of target genes and, through them, controls multiple phenotypes. The difficulties inherent in such controls cast doubt on how effective miRNAs are in driving phenotypic changes. A "simple regulation" model posits "one target-one phenotype" control under which most targeting is nonfunctional. In an alternative "coordinate regulation" model, multiple targets are assumed to control the same phenotypes coherently, and most targeting is functional. Both models have some empirical support but pose different conceptual challenges. Here, we concurrently analyze multiple targets and phenotypes associated with the miRNA-310 family (miR310s) of Drosophila Phenotypic rescue in the mir310s knockout background is achieved by promoter-directed RNA interference that restores wild-type expression. For one phenotype (eggshell morphology), we observed redundant regulation, hence rejecting "simple regulation" in favor of the "coordinate regulation" model. For other phenotypes (egg-hatching and male fertility), however, one gene shows full rescue, but three other rescues aggravate the phenotype. Overall, phenotypic controls by miR310s do not support either model. Like a thermostat that controls both heating and cooling elements to regulate temperature, redundancy and incoherence in regulation generally suggest some capacity in stability control. Our results therefore support the published view that miRNAs play a role in the canalization of transcriptome and, hence, phenotypes.
Collapse
Affiliation(s)
- Zhongqi Liufu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Yixin Zhao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Li Guo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Guangxia Miao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Juan Xiao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Yang Lyu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Yuxin Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Suhua Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Tian Tang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Chung-I Wu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
29
|
Hawley ZCE, Campos-Melo D, Droppelmann CA, Strong MJ. MotomiRs: miRNAs in Motor Neuron Function and Disease. Front Mol Neurosci 2017; 10:127. [PMID: 28522960 PMCID: PMC5415563 DOI: 10.3389/fnmol.2017.00127] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
MiRNAs are key regulators of the mammalian transcriptome that have been increasingly linked to degenerative diseases of the motor neurons. Although many of the miRNAs currently incriminated as participants in the pathogenesis of these diseases are also important to the normal development and function of motor neurons, at present there is no knowledge of the complete miRNA profile of motor neurons. In this review, we examine the current understanding with respect to miRNAs that are specifically required for motor neuron development, function and viability, and provide evidence that these should be considered as a functional network of miRNAs which we have collectively termed MotomiRs. We will also summarize those MotomiRs currently known to be associated with both amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), and discuss their potential use as biomarkers.
Collapse
Affiliation(s)
- Zachary C E Hawley
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Danae Campos-Melo
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Cristian A Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - Michael J Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada.,Department of Pathology, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada.,Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| |
Collapse
|
30
|
Pervasive Behavioral Effects of MicroRNA Regulation in Drosophila. Genetics 2017; 206:1535-1548. [PMID: 28468905 PMCID: PMC5500149 DOI: 10.1534/genetics.116.195776] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 04/23/2017] [Indexed: 11/18/2022] Open
Abstract
Picao-Osorio et al. reveal pervasive effects of microRNA regulation on complex locomotor behaviors in Drosophila larvae: over 40% of microRNAs display... The effects of microRNA (miRNA) regulation on the genetic programs underlying behavior remain largely unexplored. Despite this, recent work in Drosophila shows that mutation of a single miRNA locus (miR-iab4/iab8) affects the capacity of the larva to correct its orientation if turned upside down (self-righting, SR), suggesting that other miRNAs might also be involved in behavioral control. Here we explore this possibility, studying early larval SR behavior in a collection of 81 Drosophila miRNA mutants covering almost the entire miRNA complement of the late embryo. Unexpectedly, we observe that >40% of all miRNAs tested significantly affect SR time, revealing pervasive behavioral effects of miRNA regulation in the early larva. Detailed analyses of those miRNAs affecting SR behavior (SR-miRNAs) show that individual miRNAs can affect movement in different ways, suggesting that specific molecular and cellular elements are affected by individual miRNA mutations. Furthermore, gene expression analysis shows that the Hox gene Abdominal-B (Abd-B) represents one of the targets deregulated by several SR-miRNAs. Our work thus reveals pervasive effects of miRNA regulation on a complex innate behavior in Drosophila and suggests that miRNAs may be core components of the genetic programs underlying behavioral control in other animals too.
Collapse
|
31
|
MicroRNA-92a is a circadian modulator of neuronal excitability in Drosophila. Nat Commun 2017; 8:14707. [PMID: 28276426 PMCID: PMC5347142 DOI: 10.1038/ncomms14707] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/24/2017] [Indexed: 01/01/2023] Open
Abstract
Many biological and behavioural processes of animals are governed by an endogenous circadian clock, which is dependent on transcriptional regulation. Here we address post-transcriptional regulation and the role of miRNAs in Drosophila circadian rhythms. At least six miRNAs show cycling expression levels within the pigment dispersing factor (PDF) cell-pacemaker neurons; only mir-92a peaks during the night. In vivo calcium monitoring, dynamics of PDF projections, ArcLight, GCaMP6 imaging and sleep assays indicate that mir-92a suppresses neuronal excitability. In addition, mir-92a levels within PDF cells respond to light pulses and also affect the phase shift response. Translating ribosome affinity purification (TRAP) and in vitro luciferase reporter assay indicate that mir-92a suppresses expression of sirt2, which is homologous to human sir2 and sirt3. sirt2 RNAi also phenocopies mir-92a overexpression. These experiments indicate that sirt2 is a functional mir-92a target and that mir-92a modulates PDF neuronal excitability via suppressing SIRT2 levels in a rhythmic manner. Accumulating evidence suggests that microRNAs play a role in circadian regulation. Here the authors show that in the Drosophila brain, mir-92a suppresses the excitability of PDF neurons—key circadian pacemaker cells in Drosophila—via inhibiting the translation of its target sirt2.
Collapse
|
32
|
Li Y, Li S, Li R, Xu J, Jin P, Chen L, Ma F. Genome-wide miRNA screening reveals miR-310 family members negatively regulate the immune response in Drosophila melanogaster via co-targeting Drosomycin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 68:34-45. [PMID: 27871832 DOI: 10.1016/j.dci.2016.11.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 11/17/2016] [Accepted: 11/17/2016] [Indexed: 06/06/2023]
Abstract
Although innate immunity mediated by Toll signaling has been extensively studied in Drosophila melanogaster, the role of miRNAs in regulating the Toll-mediated immune response remains largely unknown. In this study, following Gram-positive bacterial challenge, we identified 93 differentially expressed miRNAs via genome-wide miRNA screening. These miRNAs were regarded as immune response related (IRR). Eight miRNAs were confirmed to be involved in the Toll-mediated immune response upon Gram-positive bacterial infection through genetic screening of 41 UAS-miRNA lines covering 60 miRNAs of the 93 IRR miRNAs. Interestingly, four out of these eight miRNAs, miR-310, miR-311, miR-312 and miR-313, are clustered miRNAs and belong to the miR-310 family. These miR-310 family members were shown to target and regulate the expression of Drosomycin, an antimicrobial peptide produced by Toll signaling. Taken together, our study implies important regulatory roles of miRNAs in the Toll-mediated innate immune response of Drosophila upon Gram-positive bacterial infection.
Collapse
Affiliation(s)
- Yao Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Shengjie Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Ruimin Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Jiao Xu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Liming Chen
- The Key Laboratory of Developmental Genes and Human Disease, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China.
| |
Collapse
|
33
|
Lee D, Huang TH, De La Cruz A, Callejas A, Lois C. Methods to investigate the structure and connectivity of the nervous system. Fly (Austin) 2017; 11:224-238. [PMID: 28277925 PMCID: PMC5552278 DOI: 10.1080/19336934.2017.1295189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Understanding the computations that take place in neural circuits requires identifying how neurons in those circuits are connected to one another. In addition, recent research indicates that aberrant neuronal wiring may be the cause of several neurodevelopmental disorders, further emphasizing the importance of identifying the wiring diagrams of brain circuits. To address this issue, several new approaches have been recently developed. In this review, we describe several methods that are currently available to investigate the structure and connectivity of the brain, and discuss their strengths and limitations.
Collapse
Affiliation(s)
- Donghyung Lee
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| | - Ting-Hao Huang
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| | - Aubrie De La Cruz
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| | - Antuca Callejas
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA.,b Department of Cell Biology, School of Science , University of Extremadura , Badajoz , Spain
| | - Carlos Lois
- a Division of Biology and Biological Engineering , California Institute of Technology , Pasadena , CA , USA
| |
Collapse
|
34
|
Perry S, Kiragasi B, Dickman D, Ray A. The Role of Histone Deacetylase 6 in Synaptic Plasticity and Memory. Cell Rep 2017; 18:1337-1345. [PMID: 28178513 PMCID: PMC5387061 DOI: 10.1016/j.celrep.2017.01.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 10/28/2016] [Accepted: 01/12/2017] [Indexed: 02/02/2023] Open
Abstract
Histone deacetylases (HDACs) have been extensively studied as drug targets in neurodegenerative diseases, but less is known about their role in healthy neurons. We tested zinc-dependent HDACs using RNAi in Drosophila melanogaster and found memory deficits with RPD3 and HDAC6. We demonstrate that HDAC6 is required in both the larval and adult stages for normal olfactory memory retention. Neuronal expression of HDAC6 rescued memory deficits, and we demonstrate that the N-terminal deacetylase (DAC) domain is required for this ability. This suggests that deacetylation of synaptic targets associated with the first DAC domain, such as the active-zone scaffold Bruchpilot, is required for memory retention. Finally, electrophysiological experiments at the neuromuscular junction reveal that HDAC6 mutants exhibit a partial block of homeostatic plasticity, suggesting that HDAC6 may be required for the stabilization of synaptic strength. The learning deficit we observe in HDAC6 mutants could be a behavioral consequence of these synaptic defects.
Collapse
Affiliation(s)
- Sarah Perry
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, Riverside, CA 92521, USA
| | - Beril Kiragasi
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089, USA
| | - Anandasankar Ray
- Graduate Program in Genetics, Genomics, and Bioinformatics, University of California, Riverside, Riverside, CA 92521, USA; Center for Disease Vector Research, University of California, Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
35
|
Park A, Ghezzi A, Wijesekera TP, Atkinson NS. Genetics and genomics of alcohol responses in Drosophila. Neuropharmacology 2017; 122:22-35. [PMID: 28161376 DOI: 10.1016/j.neuropharm.2017.01.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/24/2017] [Accepted: 01/29/2017] [Indexed: 02/07/2023]
Abstract
Drosophila melanogaster has become a significant model organism for alcohol research. In flies, a rich variety of behaviors can be leveraged for identifying genes affecting alcohol responses and adaptations. Furthermore, almost all genes can be easily genetically manipulated. Despite the great evolutionary distance between flies and mammals, many of the same genes have been implicated in strikingly similar alcohol-induced behaviors. A major problem in medical research today is that it is difficult to extrapolate from any single model system to humans. Strong evolutionary conservation of a mechanistic response between distantly related organisms, such as flies and mammals, is a powerful predictor that conservation will continue all the way to humans. This review describes the state of the Drosophila alcohol research field. It describes common alcohol behavioral assays, the independent origins of resistance and tolerance, the results of classical genetic screens and candidate gene analysis, and the outcomes of recent genomics studies employing GWAS, transcriptome, miRNA, and genome-wide histone acetylation surveys. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Annie Park
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Alfredo Ghezzi
- Department of Biology, University of Puerto Rico, Rio Piedras. San Juan, PR, United States
| | - Thilini P Wijesekera
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States
| | - Nigel S Atkinson
- Department of Neuroscience and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
36
|
Ghezzi A, Zomeno M, Pietrzykowski AZ, Atkinson NS. Immediate-early alcohol-responsive miRNA expression in Drosophila. J Neurogenet 2016; 30:195-204. [PMID: 27845601 DOI: 10.1080/01677063.2016.1252764] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
At the core of the changes characteristic of alcoholism are alterations in gene expression in the brain of the addicted individual. These changes are believed to underlie some of the neuroadaptations that promote compulsive drinking. Unfortunately, the mechanisms by which alcohol consumption produces changes in gene expression remain poorly understood. MicroRNAs (miRNAs) have emerged as important regulators of gene expression because they can coordinately modulate the translation efficiency of large sets of specific mRNAs. Here, we investigate the early miRNA responses elicited by an acute sedating dose of alcohol in the Drosophila model organism. In our analysis, we combine the power of next-generation sequencing with Drosophila genetics to identify alcohol-sensitive miRNAs and to functionally test them for a role in modulating alcohol sensitivity. We identified 14 known Drosophila miRNAs, and 13 putative novel miRNAs that respond to an acute sedative exposure to alcohol. Using the GeneSwitch Gal4/UAS system, a subset of these ethanol-responsive miRNAs was functionally tested to determine their individual contribution in modulating ethanol sensitivity. We identified two microRNAs that when overexpressed significantly increased ethanol sensitivity: miR-6 and miR-310. MicroRNA target prediction analysis revealed that the different alcohol-responsive miRNAs target-overlapping sets of mRNAs. Alcoholism is the product of accumulated cellular changes produced by chronic ethanol consumption. Although all of the changes described herein are extremely rapid responses evoked by a single ethanol exposure, understanding the gene expression changes that occur in the first few minutes after ethanol exposure will help us to categorize ethanol responses into those that are near instantaneous and those that are emergent responses produced only by repeated ethanol exposure.
Collapse
Affiliation(s)
- Alfredo Ghezzi
- a Department of Biology , University of Puerto Rico , Rio Piedras, San Juan , Puerto Rico
| | - Marie Zomeno
- b Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research , The University of Texas at Austin , Austin , TX , USA
| | - Andrzej Z Pietrzykowski
- c The Biologically Inspired Neural and Dynamical Systems (BINDS) Lab, Department of Computer Science , University of Massachusetts Amherst , Amherst , MA , USA
| | - Nigel S Atkinson
- b Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research , The University of Texas at Austin , Austin , TX , USA
| |
Collapse
|
37
|
Poon VY, Gu M, Ji F, VanDongen AM, Fivaz M. miR-27b shapes the presynaptic transcriptome and influences neurotransmission by silencing the polycomb group protein Bmi1. BMC Genomics 2016; 17:777. [PMID: 27716060 PMCID: PMC5050705 DOI: 10.1186/s12864-016-3139-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 09/28/2016] [Indexed: 12/02/2022] Open
Abstract
Background MicroRNAs (miRNAs) are short non-coding RNAs that are emerging as important post-transcriptional regulators of neuronal and synaptic development. The precise impact of miRNAs on presynaptic function and neurotransmission remains, however, poorly understood. Results Here, we identify miR-27b—an abundant neuronal miRNA implicated in neurological disorders—as a global regulator of the presynaptic transcriptome. miR-27b influences the expression of three quarters of genes associated with presynaptic function in cortical neurons. Contrary to expectation, a large majority of these genes are up-regulated by miR-27b. This stimulatory effect is mediated by miR-27b-directed silencing of several transcriptional repressors that cooperate to suppress the presynaptic transcriptome. The strongest repressive activity appears to be mediated by Bmi1, a component of the polycomb repressive complex implicated in self-renewal of neural stem cells. miR-27b knockdown leads to reduced synaptogenesis and to a marked decrease in neural network activity, which is fully restored by RNAi-mediated silencing of Bmi1. Conclusions We conclude that silencing of Bmi1 by miR-27b relieves repression of the presynaptic transcriptome and supports neurotransmission in cortical networks. These results expand the repressive activity of Bmi1 to genes involved in synaptic function and identify a unique post-transcriptional circuitry that stimulates expression of synaptic genes and promotes synapse differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3139-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivian Y Poon
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Minxia Gu
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Fang Ji
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Antonius M VanDongen
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Marc Fivaz
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
38
|
Fernandes D, Carvalho AL. Mechanisms of homeostatic plasticity in the excitatory synapse. J Neurochem 2016; 139:973-996. [PMID: 27241695 DOI: 10.1111/jnc.13687] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/30/2022]
Abstract
Brain development, sensory information processing, and learning and memory processes depend on Hebbian forms of synaptic plasticity, and on the remodeling and pruning of synaptic connections. Neurons in networks implicated in these processes carry out their functions while facing constant perturbation; homeostatic responses are therefore required to maintain neuronal activity within functional ranges for proper brain function. Here, we will review in vitro and in vivo studies demonstrating that several mechanisms underlie homeostatic plasticity of excitatory synapses, and identifying participant molecular players. Emerging evidence suggests a link between disrupted homeostatic synaptic plasticity and neuropsychiatric and neurologic disorders. Hebbian forms of synaptic plasticity, such as long-term potentiation (LTP), induce long-lasting changes in synaptic strength, which can be destabilizing and drive activity to saturation. Conversely, homeostatic plasticity operates to compensate for prolonged activity changes, stabilizing neuronal firing within a dynamic physiological range. We review mechanisms underlying homeostatic plasticity, and address how neurons integrate distinct forms of plasticity for proper brain function. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Dominique Fernandes
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,PDBEB-Doctoral Program in Experimental Biology and Biomedicine, Interdisciplinary Research Institute (III-UC), University of Coimbra, Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
39
|
Hedgehog Signaling Strength Is Orchestrated by the mir-310 Cluster of MicroRNAs in Response to Diet. Genetics 2016; 202:1167-83. [PMID: 26801178 PMCID: PMC4788116 DOI: 10.1534/genetics.115.185371] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/18/2016] [Indexed: 01/08/2023] Open
Abstract
Since the discovery of microRNAs (miRNAs) only two decades ago, they have emerged as an essential component of the gene regulatory machinery. miRNAs have seemingly paradoxical features: a single miRNA is able to simultaneously target hundreds of genes, while its presence is mostly dispensable for animal viability under normal conditions. It is known that miRNAs act as stress response factors; however, it remains challenging to determine their relevant targets and the conditions under which they function. To address this challenge, we propose a new workflow for miRNA function analysis, by which we found that the evolutionarily young miRNA family, the mir-310s (mir-310/mir-311/mir-312/mir-313), are important regulators of Drosophila metabolic status. mir-310s-deficient animals have an abnormal diet-dependent expression profile for numerous diet-sensitive components, accumulate fats, and show various physiological defects. We found that the mir-310s simultaneously repress the production of several regulatory factors (Rab23, DHR96, and Ttk) of the evolutionarily conserved Hedgehog (Hh) pathway to sharpen dietary response. As the mir-310s expression is highly dynamic and nutrition sensitive, this signal relay model helps to explain the molecular mechanism governing quick and robust Hh signaling responses to nutritional changes. Additionally, we discovered a new component of the Hh signaling pathway in Drosophila, Rab23, which cell autonomously regulates Hh ligand trafficking in the germline stem cell niche. How organisms adjust to dietary fluctuations to sustain healthy homeostasis is an intriguing research topic. These data are the first to report that miRNAs can act as executives that transduce nutritional signals to an essential signaling pathway. This suggests miRNAs as plausible therapeutic agents that can be used in combination with low calorie and cholesterol diets to manage quick and precise tissue-specific responses to nutritional changes.
Collapse
|
40
|
Abstract
MicroRNAs are short noncoding, ~22-nucleotide RNAs that regulate protein abundance. The growth in our understanding of this class of RNAs has been rapid since their discovery just over a decade ago. We now appreciate that miRNAs are deeply embedded within the genetic networks that control basic features of metazoan cells including their identity, metabolism, and reproduction. The Drosophila melanogaster model system has made and will continue to make important contributions to this analysis. Intended as an introductory overview, here we review the current methods and resources available for functional analysis of fly miRNAs for those interested in performing this type of analysis.
Collapse
Affiliation(s)
- Geetanjali Chawla
- Department of Biology, Indiana University, Jordan Hall, 1001 East Third St., Bloomington, IN, 47405, USA
| | - Arthur Luhur
- Department of Biology, Indiana University, Jordan Hall, 1001 East Third St., Bloomington, IN, 47405, USA
| | - Nicholas Sokol
- Department of Biology, Indiana University, Jordan Hall, 1001 East Third St., Bloomington, IN, 47405, USA.
| |
Collapse
|
41
|
Ninova M, Ronshaugen M, Griffiths-Jones S. MicroRNA evolution, expression, and function during short germband development in Tribolium castaneum. Genome Res 2015; 26:85-96. [PMID: 26518483 PMCID: PMC4691753 DOI: 10.1101/gr.193367.115] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/20/2015] [Indexed: 01/12/2023]
Abstract
MicroRNAs are well-established players in the development of multicellular animals. Most of our understanding of microRNA function in arthropod development comes from studies in Drosophila. Despite their advantages as model systems, the long germband embryogenesis of fruit flies is an evolutionary derived state restricted to several holometabolous insect lineages. MicroRNA evolution and expression across development in animals exhibiting the ancestral and more widespread short germband mode of embryogenesis has not been characterized. We sequenced small RNA libraries of oocytes and successive intervals covering the embryonic development of the short germband model organism, Tribolium castaneum. We analyzed the evolution and temporal expression of the microRNA complement and sequenced libraries of total RNA to investigate the relationships with microRNA target expression. We show microRNA maternal loading and sequence-specific 3′ end nontemplate oligoadenylation of maternally deposited microRNAs that is conserved between Tribolium and Drosophila. We further uncover large clusters encoding multiple paralogs from several Tribolium-specific microRNA families expressed during a narrow interval of time immediately after the activation of zygotic transcription. These novel microRNAs, together with several early expressed conserved microRNAs, target a significant number of maternally deposited transcripts. Comparison with Drosophila shows that microRNA-mediated maternal transcript targeting is a conserved process in insects, but the number and sequences of microRNAs involved have diverged. The expression of fast-evolving and species-specific microRNAs in the early blastoderm of T. castaneum is consistent with previous findings in Drosophila and shows that the unique permissiveness for microRNA innovation at this stage is a conserved phenomenon.
Collapse
Affiliation(s)
- Maria Ninova
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Matthew Ronshaugen
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Sam Griffiths-Jones
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
42
|
Sanchez-Díaz I, Rosales-Bravo F, Reyes-Taboada JL, Covarrubias AA, Narvaez-Padilla V, Reynaud E. The Esg Gene Is Involved in Nicotine Sensitivity in Drosophila melanogaster. PLoS One 2015; 10:e0133956. [PMID: 26222315 PMCID: PMC4519288 DOI: 10.1371/journal.pone.0133956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 07/03/2015] [Indexed: 12/04/2022] Open
Abstract
In humans, there is a strong correlation between sensitivity to substances of abuse and addiction risk. This differential tolerance to drugs has a strong genetic component. The identification of human genetic factors that alter drug tolerance has been a difficult task. For this reason and taking advantage of the fact that Drosophila responds similarly to humans to many drugs, and that genetically it has a high degree of homology (sharing at least 70% of genes known to be involved in human genetic diseases), we looked for genes in Drosophila that altered their nicotine sensitivity. We developed an instantaneous nicotine vaporization technique that exposed flies in a reproducible way. The amount of nicotine sufficient to "knock out" half of control flies for 30 minutes was determined and this parameter was defined as Half Recovery Time (HRT). Two fly lines, L4 and L70, whose HRT was significantly longer than control´s were identified. The L4 insertion is a loss of function allele of the transcriptional factor escargot (esg), whereas L70 insertion causes miss-expression of the microRNA cluster miR-310-311-312-313 (miR-310c). In this work, we demonstrate that esg loss of function induces nicotine sensitivity possibly by altering development of sensory organs and neurons in the medial section of the thoracoabdominal ganglion. The ectopic expression of the miR-310c also induces nicotine sensitivity by lowering Esg levels thus disrupting sensory organs and possibly to the modulation of other miR-310c targets.
Collapse
Affiliation(s)
- Iván Sanchez-Díaz
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal, 510–3, Cuernavaca 62210, México
| | - Fernando Rosales-Bravo
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca, Morelos 62209, México
| | - José Luis Reyes-Taboada
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal, 510–3, Cuernavaca 62210, Mexico
| | - Alejandra A Covarrubias
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal, 510–3, Cuernavaca 62210, Mexico
| | - Verónica Narvaez-Padilla
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca, Morelos 62209, México
| | - Enrique Reynaud
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Avenida Universidad, 2001, Apartado Postal, 510–3, Cuernavaca 62210, México
| |
Collapse
|
43
|
Lyons PJ, Storey KB, Morin P. Expression of miRNAs in response to freezing and anoxia stresses in the freeze tolerant fly Eurosta solidaginis. Cryobiology 2015; 71:97-102. [PMID: 25998089 DOI: 10.1016/j.cryobiol.2015.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 05/11/2015] [Accepted: 05/12/2015] [Indexed: 01/15/2023]
Abstract
Insect cold hardiness is associated with substantial metabolic rate suppression, often including developmental diapause as well as metabolic suppression imposed by freezing and freeze-associated oxygen limitation. MicroRNAs, small non-coding transcripts that bind to mRNA, are known modulators of hypometabolism in freeze tolerant insects. To further contribute to the growing signature of stress-responsive miRNAs, this study amplified and quantified changes in the expression levels of four microRNA species, miR-8, miR-9, miR-92b and miR-277, in response to freezing or anoxia exposures of freeze tolerant gall fly larvae, Eurosta solidaginis. MiR-92b levels were significantly elevated by 1.57-fold in frozen E. solidaginis at -15°C as compared with 5°C controls, whereas miR-92b levels were significantly reduced in anoxic E. solidaginis to levels that were 0.77-fold as compared with larvae held under normoxic conditions. The other miRNAs investigated showed no significant changes in stressed larvae. These data demonstrate differential miR-92b expression in frozen/anoxic versus control insect larvae and position this miRNA as a stress responsive marker in this model insect.
Collapse
Affiliation(s)
- Pierre J Lyons
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine-Maillet avenue, Moncton, New Brunswick E1A 3E9, Canada
| | - Kenneth B Storey
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Pier Morin
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine-Maillet avenue, Moncton, New Brunswick E1A 3E9, Canada.
| |
Collapse
|
44
|
Gaviño MA, Ford KJ, Archila S, Davis GW. Homeostatic synaptic depression is achieved through a regulated decrease in presynaptic calcium channel abundance. eLife 2015; 4. [PMID: 25884248 PMCID: PMC4443758 DOI: 10.7554/elife.05473] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 04/16/2015] [Indexed: 11/13/2022] Open
Abstract
Homeostatic signaling stabilizes synaptic transmission at the neuromuscular junction (NMJ) of Drosophila, mice, and human. It is believed that homeostatic signaling at the NMJ is bi-directional and considerable progress has been made identifying mechanisms underlying the homeostatic potentiation of neurotransmitter release. However, very little is understood mechanistically about the opposing process, homeostatic depression, and how bi-directional plasticity is achieved. Here, we show that homeostatic potentiation and depression can be simultaneously induced, demonstrating true bi-directional plasticity. Next, we show that mutations that block homeostatic potentiation do not alter homeostatic depression, demonstrating that these are genetically separable processes. Finally, we show that homeostatic depression is achieved by decreased presynaptic calcium channel abundance and calcium influx, changes that are independent of the presynaptic action potential waveform. Thus, we identify a novel mechanism of homeostatic synaptic plasticity and propose a model that can account for the observed bi-directional, homeostatic control of presynaptic neurotransmitter release.
Collapse
Affiliation(s)
- Michael A Gaviño
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Kevin J Ford
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Santiago Archila
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
45
|
Brusich DJ, Spring AM, Frank CA. A single-cross, RNA interference-based genetic tool for examining the long-term maintenance of homeostatic plasticity. Front Cell Neurosci 2015; 9:107. [PMID: 25859184 PMCID: PMC4374470 DOI: 10.3389/fncel.2015.00107] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/09/2015] [Indexed: 11/15/2022] Open
Abstract
Homeostatic synaptic plasticity (HSP) helps neurons and synapses maintain physiologically appropriate levels of output. The fruit fly Drosophila melanogaster larval neuromuscular junction (NMJ) is a valuable model for studying HSP. Here we introduce a genetic tool that allows fruit fly researchers to examine the lifelong maintenance of HSP with a single cross. The tool is a fruit fly stock that combines the GAL4/UAS expression system with RNA interference (RNAi)-based knock down of a glutamate receptor subunit gene. With this stock, we uncover important new information about the maintenance of HSP. We address an open question about the role that presynaptic CaV2-type Ca2+ channels play in NMJ homeostasis. Published experiments have demonstrated that hypomorphic missense mutations in the CaV2 α1a subunit gene cacophony (cac) can impair homeostatic plasticity at the NMJ. Here we report that reducing cac expression levels by RNAi is not sufficient to impair homeostatic plasticity. The presence of wild-type channels appears to support HSP—even when total CaV2 function is severely reduced. We also conduct an RNAi- and electrophysiology-based screen to identify new factors required for sustained homeostatic signaling throughout development. We uncover novel roles in HSP for Drosophila homologs of Cysteine string protein (CSP) and Phospholipase Cβ (Plc21C). We characterize those roles through follow-up genetic tests. We discuss how CSP, Plc21C, and associated factors could modulate presynaptic CaV2 function, presynaptic Ca2+ handling, or other signaling processes crucial for sustained homeostatic regulation of NMJ function throughout development. Our findings expand the scope of signaling pathways and processes that contribute to the durable strength of the NMJ.
Collapse
Affiliation(s)
- Douglas J Brusich
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| | - Ashlyn M Spring
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa Iowa City, IA, USA ; Interdisciplinary Graduate Program in Genetics, University of Iowa Iowa City, IA, USA
| | - C Andrew Frank
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa Iowa City, IA, USA ; Interdisciplinary Programs in Genetics, Neuroscience, and MCB, University of Iowa Iowa City, IA, USA
| |
Collapse
|
46
|
Kirby TJ, Chaillou T, McCarthy JJ. The role of microRNAs in skeletal muscle health and disease. Front Biosci (Landmark Ed) 2015; 20:37-77. [PMID: 25553440 DOI: 10.2741/4298] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the last decade non-coding RNAs have emerged as importance regulators of gene expression. In particular, microRNAs are a class of small RNAs of ∼ 22 nucleotides that repress gene expression through a post-transcriptional mechanism. MicroRNAs have been shown to be involved in a broader range of biological processes, both physiological and pathological, including myogenesis, adaptation to exercise and various myopathies. The purpose of this review is to provide a comprehensive summary of what is currently known about the role of microRNAs in skeletal muscle health and disease.
Collapse
Affiliation(s)
- Tyler J Kirby
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Thomas Chaillou
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, KY, USA, 2Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
47
|
Abstract
It is well established that the active properties of nerve and muscle cells are stabilized by homeostatic signaling systems. In organisms ranging from Drosophila to humans, neurons restore baseline function in the continued presence of destabilizing perturbations by rebalancing ion channel expression, modifying neurotransmitter receptor surface expression and trafficking, and modulating neurotransmitter release. This review focuses on the homeostatic modulation of presynaptic neurotransmitter release, termed presynaptic homeostasis. First, we highlight criteria that can be used to define a process as being under homeostatic control. Next, we review the remarkable conservation of presynaptic homeostasis at the Drosophila, mouse, and human neuromuscular junctions and emerging parallels at synaptic connections in the mammalian central nervous system. We then highlight recent progress identifying cellular and molecular mechanisms. We conclude by reviewing emerging parallels between the mechanisms of homeostatic signaling and genetic links to neurological disease.
Collapse
Affiliation(s)
- Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158;
| | | |
Collapse
|
48
|
miRNA-based buffering of the cobblestone-lissencephaly-associated extracellular matrix receptor dystroglycan via its alternative 3'-UTR. Nat Commun 2014; 5:4906. [PMID: 25232965 PMCID: PMC4199286 DOI: 10.1038/ncomms5906] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 08/02/2014] [Indexed: 11/08/2022] Open
Abstract
Many proteins are expressed dynamically during different stages of cellular life and the accuracy of protein amounts is critical for cell endurance. Therefore, cells should have a perceptive system that notifies about fluctuations in the amounts of certain components and an executive system that efficiently restores their precise levels. At least one mechanism that evolution has employed for this task is regulation of 3'-UTR length for microRNA targeting. Here we show that in Drosophila the microRNA complex miR-310s acts as an executive mechanism to buffer levels of the muscular dystrophy-associated extracellular matrix receptor dystroglycan via its alternative 3'-UTR. miR-310s gene expression fluctuates depending on dystroglycan amounts and nitric oxide signalling, which perceives dystroglycan levels and regulates microRNA gene expression. Aberrant levels of dystroglycan or deficiencies in miR-310s and nitric oxide signalling result in cobblestone brain appearance, resembling human lissencephaly type II phenotype.
Collapse
|
49
|
Frank CA. How voltage-gated calcium channels gate forms of homeostatic synaptic plasticity. Front Cell Neurosci 2014; 8:40. [PMID: 24592212 PMCID: PMC3924756 DOI: 10.3389/fncel.2014.00040] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/28/2014] [Indexed: 01/15/2023] Open
Abstract
Throughout life, animals face a variety of challenges such as developmental growth, the presence of toxins, or changes in temperature. Neuronal circuits and synapses respond to challenges by executing an array of neuroplasticity paradigms. Some paradigms allow neurons to up- or downregulate activity outputs, while countervailing ones ensure that outputs remain within appropriate physiological ranges. A growing body of evidence suggests that homeostatic synaptic plasticity (HSP) is critical in the latter case. Voltage-gated calcium channels gate forms of HSP. Presynaptically, the aggregate data show that when synapse activity is weakened, homeostatic signaling systems can act to correct impairments, in part by increasing calcium influx through presynaptic CaV2-type channels. Increased calcium influx is often accompanied by parallel increases in the size of active zones and the size of the readily releasable pool of presynaptic vesicles. These changes coincide with homeostatic enhancements of neurotransmitter release. Postsynaptically, there is a great deal of evidence that reduced network activity and loss of calcium influx through CaV1-type calcium channels also results in adaptive homeostatic signaling. Some adaptations drive presynaptic enhancements of vesicle pool size and turnover rate via retrograde signaling, as well as de novo insertion of postsynaptic neurotransmitter receptors. Enhanced calcium influx through CaV1 after network activation or single cell stimulation can elicit the opposite response-homeostatic depression via removal of excitatory receptors. There exist intriguing links between HSP and calcium channelopathies-such as forms of epilepsy, migraine, ataxia, and myasthenia. The episodic nature of some of these disorders suggests alternating periods of stable and unstable function. Uncovering information about how calcium channels are regulated in the context of HSP could be relevant toward understanding these and other disorders.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine Iowa City, IA, USA
| |
Collapse
|
50
|
Kye MJ, Gonçalves IDCG. The role of miRNA in motor neuron disease. Front Cell Neurosci 2014; 8:15. [PMID: 24523674 PMCID: PMC3906579 DOI: 10.3389/fncel.2014.00015] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/10/2014] [Indexed: 12/13/2022] Open
Abstract
microRNA is a subset of endogenous non-coding RNA. It binds to partially complementary sequences in mRNAs and inhibits mRNA translation by either blocking translational machinery or degrading mRNAs. It is involved in various cellular processes including cell cycle, development, metabolism, and synaptic plasticity. Dysregulation of miRNA expression and function is reported in various diseases including cancer, metabolic disorders as well as neurological disorders. In nervous system, miRNA related pathways play a very important role in development and function of neuronal cells. Moreover, numerous evidences suggest that dysregulated miRNA related pathways contribute to pathology of neurological disorders such as Alzheimer’s disease, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA). Here, we review current knowledge about the role of miRNAs in motor neuron disorders, especially about two common diseases: SMA and ALS.
Collapse
Affiliation(s)
- Min Jeong Kye
- Institute of Human Genetics, University of Cologne Cologne, Germany ; Institute for Genetics, University of Cologne Cologne, Germany
| | - Inês do Carmo G Gonçalves
- Institute of Human Genetics, University of Cologne Cologne, Germany ; Institute for Genetics, University of Cologne Cologne, Germany
| |
Collapse
|