51
|
Pratomo AR, Salim E, Hori A, Kuraishi T. Drosophila as an Animal Model for Testing Plant-Based Immunomodulators. Int J Mol Sci 2022; 23:ijms232314801. [PMID: 36499123 PMCID: PMC9735809 DOI: 10.3390/ijms232314801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Allopathic medicines play a key role in the prevention and treatment of diseases. However, long-term consumption of these medicines may cause serious undesirable effects that harm human health. Plant-based medicines have emerged as alternatives to allopathic medicines because of their rare side effects. They contain several compounds that have the potential to improve health and treat diseases in humans, including their function as immunomodulators to treat immune-related diseases. Thus, the discovery of potent and safe immunomodulators from plants is gaining considerable research interest. Recently, Drosophila has gained prominence as a model organism in evaluating the efficacy of plant and plant-derived substances. Drosophila melanogaster "fruit fly" is a well-known, high-throughput model organism that has been used to study different biological aspects of development and diseases for more than 110 years. Most developmental and cell signaling pathways and 75% of human disease-related genes are conserved between humans and Drosophila. Using Drosophila, one can easily examine the pharmacological effects of plants/plant-derived components by employing a variety of tests in flies, such as survival, anti-inflammatory, antioxidant, and cell death tests. This review focused on D. melanogaster's potential for identifying immunomodulatory features associated with plants/plant-derived components.
Collapse
Affiliation(s)
- Andre Rizky Pratomo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Emil Salim
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia
- Correspondence: (E.S.); (T.K.)
| | - Aki Hori
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
| | - Takayuki Kuraishi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-1192, Japan
- AMED-PRIME, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
- JST-FOREST, Japan Science and Technology Agency, Tokyo 102-0081, Japan
- Correspondence: (E.S.); (T.K.)
| |
Collapse
|
52
|
Multi-system responses to altered gravity and spaceflight: Insights from Drosophila melanogaster. Neurosci Biobehav Rev 2022; 142:104880. [PMID: 36126744 DOI: 10.1016/j.neubiorev.2022.104880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022]
Abstract
NASA is planning to resume human-crewed lunar missions and lay the foundation for human exploration to Mars. However, our knowledge of the overall effects of long-duration spaceflight on human physiology is limited. During spaceflight, astronauts are exposed to multiple risk factors, including gravitational changes, ionizing radiation, physiological stress, and altered circadian lighting. These factors contribute to pathophysiological responses that target different organ systems in the body. This review discusses the advancements in gravitational biology using Drosophila melanogaster, one of the first organisms to be launched into space. As a well-established spaceflight model organism, fruit flies have yielded significant information, including neurobehavioral, aging, immune, cardiovascular, developmental, and multi-omics changes across tissues and developmental stages, as detailed in this review.
Collapse
|
53
|
Cheng KC, Burdine RD, Dickinson ME, Ekker SC, Lin AY, Lloyd KCK, Lutz CM, MacRae CA, Morrison JH, O'Connor DH, Postlethwait JH, Rogers CD, Sanchez S, Simpson JH, Talbot WS, Wallace DC, Weimer JM, Bellen HJ. Promoting validation and cross-phylogenetic integration in model organism research. Dis Model Mech 2022; 15:dmm049600. [PMID: 36125045 PMCID: PMC9531892 DOI: 10.1242/dmm.049600] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Model organism (MO) research provides a basic understanding of biology and disease due to the evolutionary conservation of the molecular and cellular language of life. MOs have been used to identify and understand the function of orthologous genes, proteins, cells and tissues involved in biological processes, to develop and evaluate techniques and methods, and to perform whole-organism-based chemical screens to test drug efficacy and toxicity. However, a growing richness of datasets and the rising power of computation raise an important question: How do we maximize the value of MOs? In-depth discussions in over 50 virtual presentations organized by the National Institutes of Health across more than 10 weeks yielded important suggestions for improving the rigor, validation, reproducibility and translatability of MO research. The effort clarified challenges and opportunities for developing and integrating tools and resources. Maintenance of critical existing infrastructure and the implementation of suggested improvements will play important roles in maintaining productivity and facilitating the validation of animal models of human biology and disease.
Collapse
Affiliation(s)
- Keith C. Cheng
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
- Institute for Computational and Data Sciences, Pennsylvania State University, Park, PA 16802, USA
| | - Rebecca D. Burdine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Mary E. Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77007, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77007, USA
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55906, USA
| | - Alex Y. Lin
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - K. C. Kent Lloyd
- Mouse Biology Program, School of Medicinel, University of California Davis, Davis, CA 95618, USA
- Department of Surgery, School of Medicine, University of California Davis, Davis, CA 95618, USA
| | - Cathleen M. Lutz
- The Jackson Laboratory, Genetic Resource Science, Bar Harbor, ME 04609, USA
| | - Calum A. MacRae
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 360 Longwood Avenue, Boston, MA 02215, USA
| | - John H. Morrison
- California National Primate Research Center, University of California Davis, Davis, CA 95616, USA
- Department of Neurology, University of California Davis, Davis, CA 95616, USA
| | - David H. O'Connor
- Department of Pathology and Laboratory Medicine, University ofWisconsin-Madison, Madison, WI 53711, USA
| | | | - Crystal D. Rogers
- School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Susan Sanchez
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA
| | - Julie H. Simpson
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, CA 93117, USA
| | - William S. Talbot
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Douglas C. Wallace
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Neurological Research Institute (TCH), Baylor College of Medicine, Houston, TX 77007, USA
| |
Collapse
|
54
|
Huang G, Dierick HA. The need for unbiased genetic screens to dissect aggression in Drosophila melanogaster. Front Behav Neurosci 2022; 16:901453. [PMID: 35979224 PMCID: PMC9377312 DOI: 10.3389/fnbeh.2022.901453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Aggression is an evolutionarily conserved behavior present in most animals and is necessary for survival when competing for limited resources and mating partners. Studies have shown that aggression is modulated both genetically and epigenetically, but details of how the molecular and cellular mechanisms interact to determine aggressive behavior remain to be elucidated. In recent decades, Drosophila melanogaster has emerged as a powerful model system to understand the mechanisms that regulate aggression. Surprisingly most of the findings discovered to date have not come from genetic screens despite the fly's long and successful history of using screens to unravel its biology. Here, we highlight the tools and techniques used to successfully screen for aggression-linked behavioral elements in Drosophila and discuss the potential impact future screens have in advancing our knowledge of the underlying genetic and neural circuits governing aggression.
Collapse
Affiliation(s)
- Gary Huang
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Herman A Dierick
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
55
|
Nicotine Has a Therapeutic Window of Effectiveness in a Drosophila melanogaster Model of Parkinson’s Disease. PARKINSON'S DISEASE 2022; 2022:9291077. [PMID: 35844833 PMCID: PMC9286976 DOI: 10.1155/2022/9291077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022]
Abstract
Strong epidemiological evidence and studies in models of Parkinson's disease (PD) suggest that nicotine may be therapeutically beneficial in PD patients. However, a number of clinical trials utilizing nicotine in PD patients have had mixed results, indicating that either nicotine is not beneficial in PD patients, or an important aspect of nicotine therapy was absent. We hypothesized that nicotine must be administered early in the adult fly life in order to have beneficial effects. We show that continuous early nicotine administration improves both climbing and flight deficiencies present in homozygous park25 mutant PD model Drosophila melanogaster. Using a new climbing assay, we identify several climbing deficiencies in this PD model that are improved or rescued by continuous nicotine treatment. Amongst these benefits, it appears that nicotine improves the ability of the park25 flies to descend the climbing vial by being able to climb down more. In support of our hypothesis, we show that in order for nicotine benefits on climbing and flight to happen, nicotine administration must occur in a discrete time frame following adult fly eclosure: within one day for climbing or five days for flight. This therapeutic window of nicotine administration in this PD model fly may help to explain the lack of efficacy of nicotine in human clinical trials.
Collapse
|
56
|
Jiao W, Spreemann G, Ruchti E, Banerjee S, Vernon S, Shi Y, Stowers RS, Hess K, McCabe BD. Intact Drosophila central nervous system cellular quantitation reveals sexual dimorphism. eLife 2022; 11:74968. [PMID: 35801638 PMCID: PMC9270032 DOI: 10.7554/elife.74968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
Establishing with precision the quantity and identity of the cell types of the brain is a prerequisite for a detailed compendium of gene and protein expression in the central nervous system (CNS). Currently, however, strict quantitation of cell numbers has been achieved only for the nervous system of Caenorhabditis elegans. Here, we describe the development of a synergistic pipeline of molecular genetic, imaging, and computational technologies designed to allow high-throughput, precise quantitation with cellular resolution of reporters of gene expression in intact whole tissues with complex cellular constitutions such as the brain. We have deployed the approach to determine with exactitude the number of functional neurons and glia in the entire intact larval Drosophila CNS, revealing fewer neurons and more glial cells than previously predicted. We also discover an unexpected divergence between the sexes at this juvenile developmental stage, with the female CNS having significantly more neurons than that of males. Topological analysis of our data establishes that this sexual dimorphism extends to deeper features of CNS organisation. We additionally extended our analysis to quantitate the expression of voltage-gated potassium channel family genes throughout the CNS and uncover substantial differences in abundance. Our methodology enables robust and accurate quantification of the number and positioning of cells within intact organs, facilitating sophisticated analysis of cellular identity, diversity, and gene expression characteristics.
Collapse
Affiliation(s)
- Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - Gard Spreemann
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - Soumya Banerjee
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - Samuel Vernon
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - Ying Shi
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - R Steven Stowers
- Department of Microbiology and Cell Biology, Montana State University
| | - Kathryn Hess
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology
| |
Collapse
|
57
|
Pankau C, Cooper RL. Molecular physiology of manganese in insects. CURRENT OPINION IN INSECT SCIENCE 2022; 51:100886. [PMID: 35278758 DOI: 10.1016/j.cois.2022.100886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 06/14/2023]
Abstract
Manganese is an essential element for maintaining life. Overexposure to the metal, however, can be toxic to organisms. Given the significant function of manganese in insects, agriculture, and human disease, as well as in the healthy ecology of the planet, the biological activities of manganese in insects needs consideration. Because of the role of manganese as a cofactor for essential enzymes present in different organelles, both over and underexposure to manganese has a multifaceted effect on organisms. At the physiological level, the effects of insect exposure to the metal on enzymatic activities and consequent alteration of insect behaviors are best explained through the metal's role in modulating the dopaminergic system. Despite numerous examples that alterations in manganese homeostasis have profound effects on insects, the cellular mechanisms that ensure homeostasis of this essential metal remain presently unknown, calling for further research in this area.
Collapse
Affiliation(s)
- Cecilia Pankau
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Robin L Cooper
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
58
|
Huda A, Omelchenko AA, Vaden TJ, Castaneda AN, Ni L. Responses of different Drosophila species to temperature changes. J Exp Biol 2022; 225:275567. [PMID: 35481475 DOI: 10.1242/jeb.243708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/25/2022] [Indexed: 11/20/2022]
Abstract
Temperature is a critical environmental variable that affects the distribution, survival, and reproduction of most animals. Although temperature receptors have been identified in many animals, how these receptors respond to temperature is still unclear. Here, we describe an automated tracking method for studying the thermotactic behaviors of Drosophila larvae and adults. We build optimal experimental setups to capture behavioral recordings and analyze them using free software, Fiji and TrackMate, which do not require programming knowledge. Then, the adult thermotactic two-choice assay is applied to examine the movement and temperature preferences of nine Drosophila species. The ability or inclination to move varies among these species and at different temperatures. Distinct species prefer various ranges of temperatures. Wild-type D. melanogaster flies avoid the warmer temperature in the warm avoidance assay and the cooler temperature in the cool avoidance assay. Conversely, D. bipectinata and D. yakuba do not avoid warm or cool temperatures in the respective assays, and D. biarmipes and D. mojavensis do not avoid the warm temperature in the warm avoidance assay. These results demonstrate that Drosophila species have different mobilities and temperature preferences, which will benefit further research in exploring molecular mechanisms of temperature responsiveness.
Collapse
Affiliation(s)
- Ainul Huda
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Alisa A Omelchenko
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Thomas J Vaden
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Allison N Castaneda
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Lina Ni
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
59
|
Haynes EM, Ulland TK, Eliceiri KW. A Model of Discovery: The Role of Imaging Established and Emerging Non-mammalian Models in Neuroscience. Front Mol Neurosci 2022; 15:867010. [PMID: 35493325 PMCID: PMC9046975 DOI: 10.3389/fnmol.2022.867010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Rodents have been the dominant animal models in neurobiology and neurological disease research over the past 60 years. The prevalent use of rats and mice in neuroscience research has been driven by several key attributes including their organ physiology being more similar to humans, the availability of a broad variety of behavioral tests and genetic tools, and widely accessible reagents. However, despite the many advances in understanding neurobiology that have been achieved using rodent models, there remain key limitations in the questions that can be addressed in these and other mammalian models. In particular, in vivo imaging in mammals at the cell-resolution level remains technically difficult and demands large investments in time and cost. The simpler nervous systems of many non-mammalian models allow for precise mapping of circuits and even the whole brain with impressive subcellular resolution. The types of non-mammalian neuroscience models available spans vertebrates and non-vertebrates, so that an appropriate model for most cell biological questions in neurodegenerative disease likely exists. A push to diversify the models used in neuroscience research could help address current gaps in knowledge, complement existing rodent-based bodies of work, and bring new insight into our understanding of human disease. Moreover, there are inherent aspects of many non-mammalian models such as lifespan and tissue transparency that can make them specifically advantageous for neuroscience studies. Crispr/Cas9 gene editing and decreased cost of genome sequencing combined with advances in optical microscopy enhances the utility of new animal models to address specific questions. This review seeks to synthesize current knowledge of established and emerging non-mammalian model organisms with advances in cellular-resolution in vivo imaging techniques to suggest new approaches to understand neurodegeneration and neurobiological processes. We will summarize current tools and in vivo imaging approaches at the single cell scale that could help lead to increased consideration of non-mammalian models in neuroscience research.
Collapse
Affiliation(s)
- Elizabeth M. Haynes
- Morgridge Institute for Research, Madison, WI, United States
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, United States
| | - Tyler K. Ulland
- Department of Pathology, University of Wisconsin-Madison, Madison, WI, United States
| | - Kevin W. Eliceiri
- Morgridge Institute for Research, Madison, WI, United States
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
60
|
Salazar G, Ross G, Maserejian AE, Coutinho-Budd J. Quantifying Glial-Glial Tiling Using Automated Image Analysis in Drosophila. Front Cell Neurosci 2022; 16:826483. [PMID: 35401121 PMCID: PMC8987577 DOI: 10.3389/fncel.2022.826483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Not only do glia form close associations with neurons throughout the central nervous system (CNS), but glial cells also interact closely with other glial cells. As these cells mature, they undergo a phenomenon known as glial tiling, where they grow to abut one another, often without invading each other’s boundaries. Glial tiling occurs throughout the animal kingdom, from fruit flies to humans; however, not much is known about the glial-glial interactions that lead to and maintain this tiling. Drosophila provide a strong model to investigate glial-glial tiling, where tiling occurs both among individual glial cells of the same subtype, as well as between those of different subtypes. Furthermore, the spatial segregation of the CNS allows for the unique ability to visualize and manipulate inter-subtype interactions. Previous work in Drosophila has suggested an interaction between cortex glia and astrocytes, where astrocytes cross the normal neuropil-cortex boundary in response to dysfunctional cortex glia. Here, we further explore this interaction by implementing an automated pipeline to more fully characterize this astrocyte-cortex glial relationship. By quantifying and correlating the extent of cortex glial dysfunction and aberrant astrocyte infiltration using automated analysis, we maximize the size of the quantified dataset to reveal subtle patterns in astrocyte-cortex glial interactions. We provide a guide for creating and validating a fully-automated image analysis pipeline for exploring these interactions, and implement this pipeline to describe a significant correlation between cortex glial dysfunction and aberrant astrocyte infiltration, as well as demonstrate variations in their relationship across different regions of the CNS.
Collapse
Affiliation(s)
- Gabriela Salazar
- Department of Biology, The University of Vermont, Burlington, VT, United States.,Vermont Complex Systems Center, The University of Vermont, Burlington, VT, United States
| | - Grace Ross
- Department of Biology, The University of Vermont, Burlington, VT, United States
| | - Ariana E Maserejian
- Department of Biology, The University of Vermont, Burlington, VT, United States
| | - Jaeda Coutinho-Budd
- Department of Biology, The University of Vermont, Burlington, VT, United States
| |
Collapse
|
61
|
Disease Modeling of Rare Neurological Disorders in Zebrafish. Int J Mol Sci 2022; 23:ijms23073946. [PMID: 35409306 PMCID: PMC9000079 DOI: 10.3390/ijms23073946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Rare diseases are those which affect a small number of people compared to the general population. However, many patients with a rare disease remain undiagnosed, and a large majority of rare diseases still have no form of viable treatment. Approximately 40% of rare diseases include neurologic and neurodevelopmental disorders. In order to understand the characteristics of rare neurological disorders and identify causative genes, various model organisms have been utilized extensively. In this review, the characteristics of model organisms, such as roundworms, fruit flies, and zebrafish, are examined, with an emphasis on zebrafish disease modeling in rare neurological disorders.
Collapse
|
62
|
Honda T. Optogenetic and thermogenetic manipulation of defined neural circuits and behaviors in Drosophila. Learn Mem 2022; 29:100-109. [PMID: 35332066 PMCID: PMC8973390 DOI: 10.1101/lm.053556.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/06/2022] [Indexed: 11/25/2022]
Abstract
Neural network dynamics underlying flexible animal behaviors remain elusive. The fruit fly Drosophila melanogaster is considered an excellent model in behavioral neuroscience because of its simple neuroanatomical architecture and the availability of various genetic methods. Moreover, Drosophila larvae's transparent body allows investigators to use optical methods on freely moving animals, broadening research directions. Activating or inhibiting well-defined events in excitable cells with a fine temporal resolution using optogenetics and thermogenetics led to the association of functions of defined neural populations with specific behavioral outputs such as the induction of associative memory. Furthermore, combining optogenetics and thermogenetics with state-of-the-art approaches, including connectome mapping and machine learning-based behavioral quantification, might provide a complete view of the experience- and time-dependent variations of behavioral responses. These methodologies allow further understanding of the functional connections between neural circuits and behaviors such as chemosensory, motivational, courtship, and feeding behaviors and sleep, learning, and memory.
Collapse
Affiliation(s)
- Takato Honda
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| |
Collapse
|
63
|
Sheardown E, Mech AM, Petrazzini MEM, Leggieri A, Gidziela A, Hosseinian S, Sealy IM, Torres-Perez JV, Busch-Nentwich EM, Malanchini M, Brennan CH. Translational relevance of forward genetic screens in animal models for the study of psychiatric disease. Neurosci Biobehav Rev 2022; 135:104559. [PMID: 35124155 PMCID: PMC9016269 DOI: 10.1016/j.neubiorev.2022.104559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/10/2021] [Accepted: 02/01/2022] [Indexed: 12/16/2022]
Abstract
Psychiatric disorders represent a significant burden in our societies. Despite the convincing evidence pointing at gene and gene-environment interaction contributions, the role of genetics in the etiology of psychiatric disease is still poorly understood. Forward genetic screens in animal models have helped elucidate causal links. Here we discuss the application of mutagenesis-based forward genetic approaches in common animal model species: two invertebrates, nematodes (Caenorhabditis elegans) and fruit flies (Drosophila sp.); and two vertebrates, zebrafish (Danio rerio) and mice (Mus musculus), in relation to psychiatric disease. We also discuss the use of large scale genomic studies in human populations. Despite the advances using data from human populations, animal models coupled with next-generation sequencing strategies are still needed. Although with its own limitations, zebrafish possess characteristics that make them especially well-suited to forward genetic studies exploring the etiology of psychiatric disorders.
Collapse
Affiliation(s)
- Eva Sheardown
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Aleksandra M Mech
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | | | - Adele Leggieri
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Agnieszka Gidziela
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Saeedeh Hosseinian
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Ian M Sealy
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jose V Torres-Perez
- UK Dementia Research Institute at Imperial College London and Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Elisabeth M Busch-Nentwich
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Margherita Malanchini
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Caroline H Brennan
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK.
| |
Collapse
|
64
|
Qu S, Zhu Q, Zhou H, Gao Y, Wei Y, Ma Y, Wang Z, Sun X, Zhang L, Yang Q, Kong L, Zhang L. EasyFlyTracker: A Simple Video Tracking Python Package for Analyzing Adult Drosophila Locomotor and Sleep Activity to Facilitate Revealing the Effect of Psychiatric Drugs. Front Behav Neurosci 2022; 15:809665. [PMID: 35221942 PMCID: PMC8868375 DOI: 10.3389/fnbeh.2021.809665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanism of psychiatric drugs (stimulant and non-stimulant) is still unclear. Precision medication of psychiatric disorders faces challenges in pharmacogenetics and pharmacodynamics research due to difficulties in recruiting human subjects because of possibility of substance abuse and relatively small sample sizes. Drosophila is a powerful animal model for large-scale studies of drug effects based on the precise quantification of behavior. However, a user-friendly system for high-throughput simultaneous tracking and analysis of drug-treated individual adult flies is still lacking. It is critical to quickly setup a working environment including both the hardware and software at a reasonable cost. Thus, we have developed EasyFlyTracker, an open-source Python package that can track single fruit fly in each arena and analyze Drosophila locomotor and sleep activity based on video recording to facilitate revealing the psychiatric drug effects. The current version does not support multiple fruit fly tracking. Compared with existing software, EasyFlyTracker has the advantages of low cost, easy setup and scaling, rich statistics of movement trajectories, and compatibility with different video recording systems. Also, it accepts multiple video formats such as common MP4 and AVI formats. EasyFlyTracker provides a cross-platform and user-friendly interface combining command line and graphic configurations, which allows users to intuitively understand the process of tracking and downstream analyses and automatically generates multiple files, especially plots. Users can install EasyFlyTracker, go through tutorials, and give feedback on http://easyflytracker.cibr.ac.cn. Moreover, we tested EasyFlyTracker in a study of Drosophila melanogaster on the hyperactivity-like behavior effects of two psychiatric drugs, methylphenidate and atomoxetine, which are two commonly used drugs treating attention-deficit/hyperactivity disorder (ADHD) in human. This software has the potential to accelerate basic research on drug effect studies with fruit flies.
Collapse
Affiliation(s)
- Susu Qu
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- *Correspondence: Susu Qu,
| | - Qingjie Zhu
- Chinese Institute for Brain Research, Beijing, China
| | - Han Zhou
- Chinese Institute for Brain Research, Beijing, China
| | - Yuan Gao
- Chinese Institute for Brain Research, Beijing, China
| | - Yi Wei
- Chinese Institute for Brain Research, Beijing, China
| | - Yuan Ma
- Chinese Institute for Brain Research, Beijing, China
| | - Zhicheng Wang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Xueting Sun
- Chinese Institute for Brain Research, Beijing, China
| | - Lei Zhang
- Chinese Institute for Brain Research, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Quanjun Yang
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Lei Kong
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Li Zhang
- Chinese Institute for Brain Research, Beijing, China
- Li Zhang,
| |
Collapse
|
65
|
DiCarlo GE, Wallace MT. Modeling dopamine dysfunction in autism spectrum disorder: From invertebrates to vertebrates. Neurosci Biobehav Rev 2022; 133:104494. [PMID: 34906613 PMCID: PMC8792250 DOI: 10.1016/j.neubiorev.2021.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 02/03/2023]
Abstract
Autism Spectrum Disorder (ASD) is a highly heterogeneous neurodevelopmental disorder characterized by deficits in social communication and by patterns of restricted interests and/or repetitive behaviors. The Simons Foundation Autism Research Initiative's Human Gene and CNV Modules now list over 1000 genes implicated in ASD and over 2000 copy number variant loci reported in individuals with ASD. Given this ever-growing list of genetic changes associated with ASD, it has become evident that there is likely not a single genetic cause of this disorder nor a single neurobiological basis of this disorder. Instead, it is likely that many different neurobiological perturbations (which may represent subtypes of ASD) can result in the set of behavioral symptoms that we called ASD. One such of possible subtype of ASD may be associated with dopamine dysfunction. Precise regulation of synaptic dopamine (DA) is required for reward processing and behavioral learning, behaviors which are disrupted in ASD. Here we review evidence for DA dysfunction in ASD and in animal models of ASD. Further, we propose that these studies provide a scaffold for scientists and clinicians to consider subcategorizing the ASD diagnosis based on the genetic changes, neurobiological difference, and behavioral features identified in individuals with ASD.
Collapse
Affiliation(s)
- Gabriella E DiCarlo
- Massachusetts General Hospital, Department of Medicine, Boston, MA, United States
| | - Mark T Wallace
- Vanderbilt University Brain Institute, Nashville, TN, United States; Department of Psychology, Vanderbilt University, Nashville, TN, United States; Department of Hearing & Speech Sciences, Vanderbilt University Medical Center, Nashville, TN, United States; Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, United States.
| |
Collapse
|
66
|
Ceder MM, Fredriksson R. A phylogenetic analysis between humans and D. melanogaster: A repertoire of solute carriers in humans and flies. Gene 2022; 809:146033. [PMID: 34673204 DOI: 10.1016/j.gene.2021.146033] [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: 04/06/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/04/2022]
Abstract
The solute carrier (SLC) superfamily is the largest group of transporters in humans, with the role to transport solutes across plasma membranes. The SLCs are currently divided into 65 families with 430 members. Here, we performed a detailed mining of the SLC superfamily and the recent annotated family of "atypical" SLCs in human and D. melanogaster using Hidden Markov Models and PSI-BLAST. Our analyses identified 381 protein sequences in D. melanogaster and of those, 55 proteins have not been previously identified in flies. In total, 11 of the 65 human SLC families were found to not be conserved in flies, while a few families are highly conserved, which perhaps reflects the families' functions and roles in cellular pathways. This study provides the first collection of all SLC sequences in D. melanogaster and can serve as a SLC database to be used for classification of SLCs in other phyla.
Collapse
Affiliation(s)
- Mikaela M Ceder
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden; Sensory Circuits, Department of Neuroscience, Uppsala University, Uppsala, Sweden, Mikaela.
| | - Robert Fredriksson
- Molecular Neuropharmacology, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
67
|
Sensing microbial infections in the Drosophila melanogaster genetic model organism. Immunogenetics 2022; 74:35-62. [DOI: 10.1007/s00251-021-01239-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022]
|
68
|
Bateman JM. Mitochondrial DNA Transport in Drosophila Neurons. Methods Mol Biol 2022; 2431:409-416. [PMID: 35412289 DOI: 10.1007/978-1-0716-1990-2_21] [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: 11/28/2022]
Abstract
Mitochondria are essential organelles that generate energy and play vital roles in cellular metabolism. The small circular mitochondrial genome encodes key components of the mitochondrial respiratory apparatus. Depletion of, or mutations in mitochondrial DNA (mtDNA) cause mitochondrial dysfunction and disease. mtDNA is packaged into nucleoids, which are transported throughout the cell within mitochondria. Efficient transport of nucleoids is essential in neurons, where mitochondrial function is required locally at synapses. Here I describe methods for visualization of nucleoids in Drosophila neurons using a GFP fusion of the mitochondrial transcription factor TFAM. TFAM-GFP, together with mCherry-labeled mitochondria, was used to visualize nucleoids in fixed larval segmental nerves. I also describe how these tools can be used for live imaging of nucleoid dynamics. Using Drosophila as a model system, these methods will enable further characterization and analysis of nucleoid dynamics in neurons.
Collapse
Affiliation(s)
- Joseph M Bateman
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.
| |
Collapse
|
69
|
Voelzmann A, Sanchez-Soriano N. Drosophila Primary Neuronal Cultures as a Useful Cellular Model to Study and Image Axonal Transport. Methods Mol Biol 2022; 2431:429-449. [PMID: 35412291 DOI: 10.1007/978-1-0716-1990-2_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The use of primary neuronal cultures generated from Drosophila tissue provides a powerful model for studies of transport mechanisms. Cultured fly neurons provide similarly detailed subcellular resolution and applicability of pharmacology or fluorescent dyes as mammalian primary neurons. As an experimental advantage for the mechanistic dissection of transport, fly primary neurons can be combined with the fast and highly efficient combinatorial genetics of Drosophila, and genetic tools for the manipulation of virtually every fly gene are readily available. This strategy can be performed in parallel to in vivo transport studies to address relevance of any findings. Here we will describe the generation of primary neuronal cultures from Drosophila embryos and larvae, the use of external fluorescent dyes and genetic tools to label cargo, and the key strategies for live imaging and subsequent analysis.
Collapse
Affiliation(s)
- André Voelzmann
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| | - Natalia Sanchez-Soriano
- Department of Molecular Physiology & Cell Signalling, Institute of Systems, Molecular & Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
70
|
van Alphen B, Stewart S, Iwanaszko M, Xu F, Li K, Rozenfeld S, Ramakrishnan A, Itoh TQ, Sisobhan S, Qin Z, Lear BC, Allada R. Glial immune-related pathways mediate effects of closed head traumatic brain injury on behavior and lethality in Drosophila. PLoS Biol 2022; 20:e3001456. [PMID: 35081110 PMCID: PMC8791498 DOI: 10.1371/journal.pbio.3001456] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
In traumatic brain injury (TBI), the initial injury phase is followed by a secondary phase that contributes to neurodegeneration, yet the mechanisms leading to neuropathology in vivo remain to be elucidated. To address this question, we developed a Drosophila head-specific model for TBI termed Drosophila Closed Head Injury (dCHI), where well-controlled, nonpenetrating strikes are delivered to the head of unanesthetized flies. This assay recapitulates many TBI phenotypes, including increased mortality, impaired motor control, fragmented sleep, and increased neuronal cell death. TBI results in significant changes in the transcriptome, including up-regulation of genes encoding antimicrobial peptides (AMPs). To test the in vivo functional role of these changes, we examined TBI-dependent behavior and lethality in mutants of the master immune regulator NF-κB, important for AMP induction, and found that while sleep and motor function effects were reduced, lethality effects were enhanced. Similarly, loss of most AMP classes also renders flies susceptible to lethal TBI effects. These studies validate a new Drosophila TBI model and identify immune pathways as in vivo mediators of TBI effects.
Collapse
Affiliation(s)
- Bart van Alphen
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Samuel Stewart
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Marta Iwanaszko
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- Department of Preventive Medicine—Biostatistics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Fangke Xu
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Keyin Li
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Sydney Rozenfeld
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Anujaianthi Ramakrishnan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Taichi Q. Itoh
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Shiju Sisobhan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Zuoheng Qin
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Bridget C. Lear
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
71
|
Maiellaro I. In Vivo cAMP Dynamics in Drosophila Larval Neurons. Methods Mol Biol 2022; 2483:181-194. [PMID: 35286676 DOI: 10.1007/978-1-0716-2245-2_11] [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] [Indexed: 06/14/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) is a universal second messenger that mediates a myriad of cell functions across all kingdoms of life.The ability to monitor intracellular changes of cAMP concentration in living cells using FRET-based biosensors is proving to be of paramount importance to unraveling the sophisticated organization of cAMP signaling.Here we describe the deployment of the fruit fly Drosophila melanogaster, specifically the third instar larval stage, as an in vivo model to study the spatio-temporal dynamics of cAMP in neurons. The ubiquity of cAMP signaling and conservation of fundamental mechanisms across species ensures relevance to vertebrate neurons while providing a more structurally and ethically simple model.
Collapse
Affiliation(s)
- Isabella Maiellaro
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham, UK.
| |
Collapse
|
72
|
Ahn Y, Han SH, Kim MG, Hong KB, Kim WJ, Suh HJ, Jo K. Anti-depressant effects of ethanol extract from Cannabis sativa (hemp) seed in chlorpromazine-induced Drosophila melanogaster depression model. PHARMACEUTICAL BIOLOGY 2021; 59:998-1007. [PMID: 34362287 PMCID: PMC8354181 DOI: 10.1080/13880209.2021.1949356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
CONTEXT Depression is a severe mental illness caused by a deficiency of dopamine and serotonin. Cannabis sativa L. (Cannabaceae) has long been used to treat pain, nausea, and depression. OBJECTIVE This study investigates the anti-depressant effects of C. sativa (hemp) seed ethanol extract (HE) in chlorpromazine (CPZ)-induced Drosophila melanogaster depression model. MATERIALS AND METHODS The normal group was untreated, and the control group was treated with CPZ (0.1% of media) for 7 days. The experimental groups were treated with a single HE treatment (0.5, 1.0, and 1.5% of media) and a mixture of 0.1% CPZ and HE for 7 days. The locomotor activity, behavioural patterns, depression-related gene expression, and neurotransmitters level of flies were investigated. RESULTS The behavioural patterns of individual flies were significantly reduced with 0.1% CPZ treatment. In contrast, combination treatment of 1.5% HE and 0.1% CPZ significantly increased subjective daytime activity (p < 0.001) and behavioural factors (p < 0.001). These results correlate with increased transcript levels of dopamine (p < 0.001) and serotonin (p < 0.05) receptors and concentration of dopamine (p < 0.05), levodopa (p < 0.001), 5-HTP (p < 0.05), and serotonin (p < 0.001) compared to those in the control group. DISCUSSION AND CONCLUSIONS Collectively, HE administration alleviates depression-like symptoms by modulating the circadian rhythm-related behaviours, transcript levels of neurotransmitter receptors, and neurotransmitter levels in the CPZ-induced Drosophila model. However, additional research is needed to investigate the role of HE administration in behavioural patterns, reduction of the neurotransmitter, and signalling pathways of depression in a vertebrate model system.
Collapse
Affiliation(s)
- Yejin Ahn
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Sung Hee Han
- Institute of Human Behavior & Genetic, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Min Guk Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju, Republic of Korea
| | - Woo Jung Kim
- Biocenter, Gyeonggido Business and Science Accerlerator, Suwon, Republic of Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Kyungae Jo
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
- CONTACT Kyungae Jo Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| |
Collapse
|
73
|
Mutants of the white ABCG Transporter in Drosophila melanogaster Have Deficient Olfactory Learning and Cholesterol Homeostasis. Int J Mol Sci 2021; 22:ijms222312967. [PMID: 34884779 PMCID: PMC8657504 DOI: 10.3390/ijms222312967] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/17/2022] Open
Abstract
Drosophila's white gene encodes an ATP-binding cassette G-subfamily (ABCG) half-transporter. White is closely related to mammalian ABCG family members that function in cholesterol efflux. Mutants of white have several behavioral phenotypes that are independent of visual defects. This study characterizes a novel defect of white mutants in the acquisition of olfactory memory using the aversive olfactory conditioning paradigm. The w1118 mutants learned slower than wildtype controls, yet with additional training, they reached wildtype levels of performance. The w1118 learning phenotype is also found in the wapricot and wcoral alleles, is dominant, and is rescued by genomic white and mini-white transgenes. Reducing dietary cholesterol strongly impaired olfactory learning for wildtype controls, while w1118 mutants were resistant to this deficit. The w1118 mutants displayed higher levels of cholesterol and cholesterol esters than wildtype under this low-cholesterol diet. Increasing levels of serotonin, dopamine, or both in the white mutants significantly improved w1118 learning. However, serotonin levels were not lower in the heads of the w1118 mutants than in wildtype controls. There were also no significant differences found in synapse numbers within the w1118 brain. We propose that the w1118 learning defect may be due to inefficient biogenic amine signaling brought about by altered cholesterol homeostasis.
Collapse
|
74
|
Charlton-Perkins MA, Friedrich M, Cook TA. Semper's cells in the insect compound eye: Insights into ocular form and function. Dev Biol 2021; 479:126-138. [PMID: 34343526 PMCID: PMC8410683 DOI: 10.1016/j.ydbio.2021.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/28/2022]
Abstract
The arthropod compound eye represents one of two major eye types in the animal kingdom and has served as an essential experimental paradigm for defining fundamental mechanisms underlying sensory organ formation, function, and maintenance. One of the most distinguishing features of the compound eye is the highly regular array of lens facets that define individual eye (ommatidial) units. These lens facets are produced by a deeply conserved quartet of cuticle-secreting cells, called Semper cells (SCs). Also widely known as cone cells, SCs were originally identified for their secretion of the dioptric system, i.e. the corneal lens and underlying crystalline cones. Additionally, SCs are now known to execute a diversity of patterning and glial functions in compound eye development and maintenance. Here, we present an integrated account of our current knowledge of SC multifunctionality in the Drosophila compound eye, highlighting emerging gene regulatory modules that may drive the diverse roles for these cells. Drawing comparisons with other deeply conserved retinal glia in the vertebrate single lens eye, this discussion speaks to glial cell origins and opens new avenues for understanding sensory system support programs.
Collapse
Affiliation(s)
- Mark A Charlton-Perkins
- Department of Paediatrics, Wellcome-MRC Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, United Kingdom
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Detroit, MI, 48202, USA; Department of Ophthalmological, Visual, and Anatomical Sciences, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA
| | - Tiffany A Cook
- Department of Ophthalmological, Visual, and Anatomical Sciences, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA; Center of Molecular Medicine and Genetics, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA.
| |
Collapse
|
75
|
Krzeptowski W, Walkowicz L, Krzeptowska E, Motta E, Witek K, Szramel J, Al Abaquita T, Baster Z, Rajfur Z, Rosato E, Stratoulias V, Heino TI, Pyza EM. Mesencephalic Astrocyte-Derived Neurotrophic Factor Regulates Morphology of Pigment-Dispersing Factor-Positive Clock Neurons and Circadian Neuronal Plasticity in Drosophila melanogaster. Front Physiol 2021; 12:705183. [PMID: 34646147 PMCID: PMC8502870 DOI: 10.3389/fphys.2021.705183] [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: 05/04/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Mesencephalic Astrocyte-derived Neurotrophic Factor (MANF) is one of a few neurotrophic factors described in Drosophila melanogaster (DmMANF) but its function is still poorly characterized. In the present study we found that DmMANF is expressed in different clusters of clock neurons. In particular, the PDF-positive large (l-LNv) and small (s-LNv) ventral lateral neurons, the CRYPTOCHROME-positive dorsal lateral neurons (LNd), the group 1 dorsal neurons posterior (DN1p) and different tim-positive cells in the fly's visual system. Importantly, DmMANF expression in the ventral lateral neurons is not controlled by the clock nor it affects its molecular mechanism. However, silencing DmMANF expression in clock neurons affects the rhythm of locomotor activity in light:dark and constant darkness conditions. Such phenotypes correlate with abnormal morphology of the dorsal projections of the s-LNv and with reduced arborizations of the l-LNv in the medulla of the optic lobe. Additionally, we show that DmMANF is important for normal morphology of the L2 interneurons in the visual system and for the circadian rhythm in the topology of their dendritic tree. Our results indicate that DmMANF is important not only for the development of neurites but also for maintaining circadian plasticity of neurons.
Collapse
Affiliation(s)
- Wojciech Krzeptowski
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Lucyna Walkowicz
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Ewelina Krzeptowska
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Edyta Motta
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Kacper Witek
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Joanna Szramel
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Terence Al Abaquita
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Zbigniew Baster
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland
| | - Zenon Rajfur
- Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Kraków, Poland.,Jagiellonian Center of Biomedical Imaging, Jagiellonian University, Kraków, Poland
| | - Ezio Rosato
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Vassilis Stratoulias
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland.,Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tapio I Heino
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Elżbieta M Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland.,Jagiellonian Center of Biomedical Imaging, Jagiellonian University, Kraków, Poland
| |
Collapse
|
76
|
Almeida Machado Costa C, Wang XF, Ellsworth C, Deng WM. Polyploidy in development and tumor models in Drosophila. Semin Cancer Biol 2021; 81:106-118. [PMID: 34562587 DOI: 10.1016/j.semcancer.2021.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/03/2021] [Accepted: 09/18/2021] [Indexed: 12/26/2022]
Abstract
Polyploidy, a cell status defined as more than two sets of genomic DNA, is a conserved strategy across species that can increase cell size and biosynthetic production, but the functional aspects of polyploidy are nuanced and vary across cell types. Throughout Drosophila developmental stages (embryo, larva, pupa and adult), polyploid cells are present in numerous organs and help orchestrate development while contributing to normal growth, well-being and homeostasis of the organism. Conversely, increasing evidence has shown that polyploid cells are prevalent in Drosophila tumors and play important roles in tumor growth and invasiveness. Here, we summarize the genes and pathways involved in polyploidy during normal and tumorigenic development, the mechanisms underlying polyploidization, and the functional aspects of polyploidy in development, homeostasis and tumorigenesis in the Drosophila model.
Collapse
Affiliation(s)
- Caique Almeida Machado Costa
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States
| | - Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States
| | - Calder Ellsworth
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, United States.
| |
Collapse
|
77
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
78
|
Silvernagel MP, Ling AS, Nuyujukian P. A markerless platform for ambulatory systems neuroscience. Sci Robot 2021; 6:eabj7045. [PMID: 34516749 DOI: 10.1126/scirobotics.abj7045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Motor systems neuroscience seeks to understand how the brain controls movement. To minimize confounding variables, large-animal studies typically constrain body movement from areas not under observation, ensuring consistent, repeatable behaviors. Such studies have fueled decades of research, but they may be artificially limiting the richness of neural data observed, preventing generalization to more natural movements and settings. Neuroscience studies of unconstrained movement would capture a greater range of behavior and a more complete view of neuronal activity, but instrumenting an experimental rig suitable for large animals presents substantial engineering challenges. Here, we present a markerless, full-body motion tracking and synchronized wireless neural electrophysiology platform for large, ambulatory animals. Composed of four depth (RGB-D) cameras that provide a 360° view of a 4.5-square-meters enclosed area, this system is designed to record a diverse range of neuroethologically relevant behaviors. This platform also allows for the simultaneous acquisition of hundreds of wireless neural recording channels in multiple brain regions. As behavioral and neuronal data are generated at rates below 200 megabytes per second, a single desktop can facilitate hours of continuous recording. This setup is designed for systems neuroscience and neuroengineering research, where synchronized kinematic behavior and neural data are the foundation for investigation. By enabling the study of previously unexplored movement tasks, this system can generate insights into the functioning of the mammalian motor system and provide a platform to develop brain-machine interfaces for unconstrained applications.
Collapse
Affiliation(s)
| | - Alissa S Ling
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Paul Nuyujukian
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.,Department of Bioengineering, Stanford University, Stanford, CA, USA.,Department of Neurosurgery, Stanford University, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Stanford Bio-X, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
79
|
|
80
|
Finet C, Kassner VA, Carvalho AB, Chung H, Day JP, Day S, Delaney EK, De Ré FC, Dufour HD, Dupim E, Izumitani HF, Gautério TB, Justen J, Katoh T, Kopp A, Koshikawa S, Longdon B, Loreto EL, Nunes MDS, Raja KKB, Rebeiz M, Ritchie MG, Saakyan G, Sneddon T, Teramoto M, Tyukmaeva V, Vanderlinde T, Wey EE, Werner T, Williams TM, Robe LJ, Toda MJ, Marlétaz F. DrosoPhyla: Resources for Drosophilid Phylogeny and Systematics. Genome Biol Evol 2021; 13:evab179. [PMID: 34343293 PMCID: PMC8382681 DOI: 10.1093/gbe/evab179] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2021] [Indexed: 02/06/2023] Open
Abstract
The vinegar fly Drosophila melanogaster is a pivotal model for invertebrate development, genetics, physiology, neuroscience, and disease. The whole family Drosophilidae, which contains over 4,400 species, offers a plethora of cases for comparative and evolutionary studies. Despite a long history of phylogenetic inference, many relationships remain unresolved among the genera, subgenera, and species groups in the Drosophilidae. To clarify these relationships, we first developed a set of new genomic markers and assembled a multilocus data set of 17 genes from 704 species of Drosophilidae. We then inferred a species tree with highly supported groups for this family. Additionally, we were able to determine the phylogenetic position of some previously unplaced species. These results establish a new framework for investigating the evolution of traits in fruit flies, as well as valuable resources for systematics.
Collapse
Affiliation(s)
- Cédric Finet
- Howard Hughes Medical Institute and Laboratory of Molecular Biology, University of Wisconsin, Madison, USA
| | - Victoria A Kassner
- Howard Hughes Medical Institute and Laboratory of Molecular Biology, University of Wisconsin, Madison, USA
| | - Antonio B Carvalho
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Henry Chung
- Department of Entomology, Michigan State University, USA
| | - Jonathan P Day
- Department of Genetics, University of Cambridge, United Kingdom
| | - Stephanie Day
- Department of Biological Sciences, University of Pittsburgh, USA
| | - Emily K Delaney
- Department of Evolution and Ecology, University of California-Davis, USA
| | - Francine C De Ré
- Programa de Pós-Graduação em Biodiversidade Animal, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | - Héloïse D Dufour
- Howard Hughes Medical Institute and Laboratory of Molecular Biology, University of Wisconsin, Madison, USA
| | - Eduardo Dupim
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Hiroyuki F Izumitani
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Thaísa B Gautério
- Programa de Pós-Graduação em Biologia de Ambientes Aquáticos Continentais, Universidade Federal do Rio Grande, Rio Grande do Sul, Brazil
| | - Jessa Justen
- Howard Hughes Medical Institute and Laboratory of Molecular Biology, University of Wisconsin, Madison, USA
| | - Toru Katoh
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Artyom Kopp
- Department of Evolution and Ecology, University of California-Davis, USA
| | - Shigeyuki Koshikawa
- The Hakubi Center for Advanced Research and Graduate School of Science, Kyoto University, Japan
| | - Ben Longdon
- Centre for Ecology and Conservation, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Elgion L Loreto
- Programa de Pós-Graduação em Biodiversidade Animal, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | - Maria D S Nunes
- Department of Biological and Medical Sciences, Oxford Brookes University, United Kingdom
- Centre for Functional Genomics, Oxford Brookes University, United Kingdom
| | - Komal K B Raja
- Department of Biological Sciences, Michigan Technological University, USA
| | - Mark Rebeiz
- Department of Biological Sciences, University of Pittsburgh, USA
| | | | - Gayane Saakyan
- Department of Evolution and Ecology, University of California-Davis, USA
| | - Tanya Sneddon
- School of Biology, University of St Andrews, United Kingdom
| | - Machiko Teramoto
- The Hakubi Center for Advanced Research and Graduate School of Science, Kyoto University, Japan
| | | | - Thyago Vanderlinde
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Brazil
| | - Emily E Wey
- Department of Biology, University of Dayton, USA
| | - Thomas Werner
- Department of Biological Sciences, Michigan Technological University, USA
| | | | - Lizandra J Robe
- Programa de Pós-Graduação em Biodiversidade Animal, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Biologia de Ambientes Aquáticos Continentais, Universidade Federal do Rio Grande, Rio Grande do Sul, Brazil
| | - Masanori J Toda
- Hokkaido University Museum, Hokkaido University, Sapporo, Japan
| | - Ferdinand Marlétaz
- Centre for Life’s Origins and Evolution, Department of Genetics, Evolution and Environment, University College London, United Kingdom
| |
Collapse
|
81
|
Inal MA, Bui KC, Marar A, Li S, Kner P, Kamiyama D. Imaging of In Vitro and In Vivo Neurons in Drosophila Using Stochastic Optical Reconstruction Microscopy. Curr Protoc 2021; 1:e203. [PMID: 34289261 DOI: 10.1002/cpz1.203] [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: 11/11/2022]
Abstract
The Drosophila melanogaster brain comprises different neuronal cell types that interconnect with precise patterns of synaptic connections. These patterns are essential for the normal function of the brain. To understand the connectivity patterns requires characterizing them at single-cell resolution, for which a fluorescence microscope becomes an indispensable tool. Additionally, because the neurons connect at the nanoscale, the investigation often demands super-resolution microscopy. Here, we adopt one super-resolution microscopy technique, called stochastic optical reconstruction microscopy (STORM), improving the lateral and axial resolution to ∼20 nm. This article extensively describes our methods along with considerations for sample preparation of neurons in vitro and in vivo, conjugation of dyes to antibodies, immunofluorescence labeling, and acquisition and processing of STORM data. With these tools and techniques, we open up the potential to investigate cell-cell interactions using STORM in the Drosophila nervous system. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Preparation of Drosophila primary neuronal culture and embryonic fillets Basic Protocol 2: Immunofluorescence labeling of samples Basic Protocol 3: Single-molecule fluorescence imaging Basic Protocol 4: Localization and visualization of single-molecule data Supporting Protocol: Conjugation of antibodies with STORM-compatible dyes.
Collapse
Affiliation(s)
- Melissa Ana Inal
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Kathy Clara Bui
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Abhijit Marar
- School of Electrical and Computer Engineering, University of Georgia, Athens, Georgia
| | - Shaoheng Li
- School of Electrical and Computer Engineering, University of Georgia, Athens, Georgia
| | - Peter Kner
- School of Electrical and Computer Engineering, University of Georgia, Athens, Georgia
| | - Daichi Kamiyama
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| |
Collapse
|
82
|
The early Drosophila embryo as a model system for quantitative biology. Cells Dev 2021; 168:203722. [PMID: 34298230 DOI: 10.1016/j.cdev.2021.203722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/03/2021] [Accepted: 07/13/2021] [Indexed: 11/20/2022]
Abstract
With the rise of new tools, from controlled genetic manipulations and optogenetics to improved microscopy, it is now possible to make clear, quantitative and reproducible measurements of biological processes. The humble fruit fly Drosophila melanogaster, with its ease of genetic manipulation combined with excellent imaging accessibility, has become a major model system for performing quantitative in vivo measurements. Such measurements are driving a new wave of interest from physicists and engineers, who are developing a range of testable dynamic models of active systems to understand fundamental biological processes. The reproducibility of the early Drosophila embryo has been crucial for understanding how biological systems are robust to unavoidable noise during development. Insights from quantitative in vivo experiments in the Drosophila embryo are having an impact on our understanding of critical biological processes, such as how cells make decisions and how complex tissue shape emerges. Here, to highlight the power of using Drosophila embryogenesis for quantitative biology, I focus on three main areas: (1) formation and robustness of morphogen gradients; (2) how gene regulatory networks ensure precise boundary formation; and (3) how mechanical interactions drive packing and tissue folding. I further discuss how such data has driven advances in modelling.
Collapse
|
83
|
Loesche F, Reiser MB. An Inexpensive, High-Precision, Modular Spherical Treadmill Setup Optimized for Drosophila Experiments. Front Behav Neurosci 2021; 15:689573. [PMID: 34335199 PMCID: PMC8322621 DOI: 10.3389/fnbeh.2021.689573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/04/2021] [Indexed: 11/24/2022] Open
Abstract
To pursue a more mechanistic understanding of the neural control of behavior, many neuroethologists study animal behavior in controlled laboratory environments. One popular approach is to measure the movements of restrained animals while presenting controlled sensory stimulation. This approach is especially powerful when applied to genetic model organisms, such as Drosophila melanogaster, where modern genetic tools enable unprecedented access to the nervous system for activity monitoring or targeted manipulation. While there is a long history of measuring the behavior of body- and head-fixed insects walking on an air-supported ball, the methods typically require complex setups with many custom components. Here we present a compact, simplified setup for these experiments that achieves high-performance at low cost. The simplified setup integrates existing hardware and software solutions with new component designs. We replaced expensive optomechanical and custom machined components with off-the-shelf and 3D-printed parts, and built the system around a low-cost camera that achieves 180 Hz imaging and an inexpensive tablet computer to present view-angle-corrected stimuli updated through a local network. We quantify the performance of the integrated system and characterize the visually guided behavior of flies in response to a range of visual stimuli. In this paper, we thoroughly document the improved system; the accompanying repository incorporates CAD files, parts lists, source code, and detailed instructions. We detail a complete ~$300 system, including a cold-anesthesia tethering stage, that is ideal for hands-on teaching laboratories. This represents a nearly 50-fold cost reduction as compared to a typical system used in research laboratories, yet is fully featured and yields excellent performance. We report the current state of this system, which started with a 1-day teaching lab for which we built seven parallel setups and continues toward a setup in our lab for larger-scale analysis of visual-motor behavior in flies. Because of the simplicity, compactness, and low cost of this system, we believe that high-performance measurements of tethered insect behavior should now be widely accessible and suitable for integration into many systems. This access enables broad opportunities for comparative work across labs, species, and behavioral paradigms.
Collapse
Affiliation(s)
- Frank Loesche
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Michael B Reiser
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| |
Collapse
|
84
|
Byrns CN, Saikumar J, Bonini NM. Glial AP1 is activated with aging and accelerated by traumatic brain injury. NATURE AGING 2021; 1:585-597. [PMID: 34723199 PMCID: PMC8553014 DOI: 10.1038/s43587-021-00072-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/30/2021] [Indexed: 01/05/2023]
Abstract
The emergence of degenerative disease after traumatic brain injury is often described as an acceleration of normal age-related processes. Whether similar molecular processes occur after injury and in age is unclear. Here we identify a functionally dynamic and lasting transcriptional response in glia, mediated by the conserved transcription factor AP1. In the early post-TBI period, glial AP1 is essential for recovery, ensuring brain integrity and animal survival. In sharp contrast, chronic AP1 activation promotes human tau pathology, tissue loss, and mortality. We show a similar process activates in healthy fly brains with age. In humans, AP1 activity is detected after moderate TBI and correlates with microglial activation and tau pathology. Our data provide key molecular insight into glia, highlighting that the same molecular process drives dynamic and contradictory glia behavior in TBI, and possibly age, first acting to protect but chronically promoting disease.
Collapse
Affiliation(s)
- China N Byrns
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janani Saikumar
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
85
|
Pandolfi M, Scaia MF, Fernandez MP. Sexual Dimorphism in Aggression: Sex-Specific Fighting Strategies Across Species. Front Behav Neurosci 2021; 15:659615. [PMID: 34262439 PMCID: PMC8273308 DOI: 10.3389/fnbeh.2021.659615] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Aggressive behavior is thought to have evolved as a strategy for gaining access to resources such as territory, food, and potential mates. Across species, secondary sexual characteristics such as competitive aggression and territoriality are considered male-specific behaviors. However, although female–female aggression is often a behavior that is displayed almost exclusively to protect the offspring, multiple examples of female–female competitive aggression have been reported in both invertebrate and vertebrate species. Moreover, cases of intersexual aggression have been observed in a variety of species. Genetically tractable model systems such as mice, zebrafish, and fruit flies have proven extremely valuable for studying the underlying neuronal circuitry and the genetic architecture of aggressive behavior under laboratory conditions. However, most studies lack ethological or ecological perspectives and the behavioral patterns available are limited. The goal of this review is to discuss each of these forms of aggression, male intrasexual aggression, intersexual aggression and female intrasexual aggression in the context of the most common genetic animal models and discuss examples of these behaviors in other species.
Collapse
Affiliation(s)
- Matias Pandolfi
- Department of Biodiversity and Experimental Biology, University of Buenos Aires, Buenos Aires, Argentina
| | - Maria Florencia Scaia
- Department of Biodiversity and Experimental Biology, University of Buenos Aires, Buenos Aires, Argentina
| | - Maria Paz Fernandez
- Department of Neuroscience and Behavior, Barnard College of Columbia University, New York, NY, United States
| |
Collapse
|
86
|
Lucas C, Ben-Shahar Y. The foraging gene as a modulator of division of labour in social insects. J Neurogenet 2021; 35:168-178. [PMID: 34151702 DOI: 10.1080/01677063.2021.1940173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The social ants, bees, wasps, and termites include some of the most ecologically-successful groups of animal species. Their dominance in most terrestrial environments is attributed to their social lifestyle, which enable their colonies to exploit environmental resources with remarkable efficiency. One key attribute of social insect colonies is the division of labour that emerges among the sterile workers, which represent the majority of colony members. Studies of the mechanisms that drive division of labour systems across diverse social species have provided fundamental insights into the developmental, physiological, molecular, and genomic processes that regulate sociality, and the possible genetic routes that may have led to its evolution from a solitary ancestor. Here we specifically discuss the conserved role of the foraging gene, which encodes a cGMP-dependent protein kinase (PKG). Originally identified as a behaviourally polymorphic gene that drives alternative foraging strategies in the fruit fly Drosophila melanogaster, changes in foraging expression and kinase activity were later shown to play a key role in the division of labour in diverse social insect species as well. In particular, foraging appears to regulate worker transitions between behavioural tasks and specific behavioural traits associated with morphological castes. Although the specific neuroethological role of foraging in the insect brain remains mostly unknown, studies in genetically tractable insect species indicate that PKG signalling plays a conserved role in the neuronal plasticity of sensory, cognitive and motor functions, which underlie behaviours relevant to division of labour, including appetitive learning, aggression, stress response, phototaxis, and the response to pheromones.
Collapse
Affiliation(s)
- Christophe Lucas
- Institut de Recherche sur la Biologie de l'Insecte (UMR7261), CNRS - University of Tours, Tours, France
| | - Yehuda Ben-Shahar
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
87
|
Crocker KL, Marischuk K, Rimkus SA, Zhou H, Yin JCP, Boekhoff-Falk G. Neurogenesis in the adult Drosophila brain. Genetics 2021; 219:6297258. [PMID: 34117750 PMCID: PMC8860384 DOI: 10.1093/genetics/iyab092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/08/2021] [Indexed: 11/14/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer's and Parkinson's currently affect ∼25 million people worldwide (Erkkinen et al. 2018). The global incidence of traumatic brain injury (TBI) is estimated at ∼70 million/year (Dewan et al. 2018). Both neurodegenerative diseases and TBI remain without effective treatments. We are utilizing adult Drosophila melanogaster to investigate the mechanisms of brain regeneration with the long term goal of identifying targets for neural regenerative therapies. We specifically focused on neurogenesis, i.e. the generation of new cells, as opposed to the regrowth of specific subcellular structures such as axons. Like mammals, Drosophila have few proliferating cells in the adult brain. Nonetheless, within 24 hours of a Penetrating Traumatic Brain Injury (PTBI) to the central brain, there is a significant increase in the number of proliferating cells. We subsequently detect both new glia and new neurons and the formation of new axon tracts that target appropriate brain regions. Glial cells divide rapidly upon injury to give rise to new glial cells. Other cells near the injury site upregulate neural progenitor genes including asense and deadpan and later give rise to the new neurons. Locomotor abnormalities observed after PTBI are reversed within two weeks of injury, supporting the idea that there is functional recovery. Together, these data indicate that adult Drosophila brains are capable of neuronal repair. We anticipate that this paradigm will facilitate the dissection of the mechanisms of neural regeneration and that these processes will be relevant to human brain repair.
Collapse
Affiliation(s)
- Kassi L Crocker
- Genetics Graduate Training Program, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA.,Science and Medicine Graduate Research Scholars Program, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Khailee Marischuk
- Genetics Graduate Training Program, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Stacey A Rimkus
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Hong Zhou
- Department of Genetics, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Jerry C P Yin
- Department of Genetics, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Grace Boekhoff-Falk
- Genetics Graduate Training Program, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
88
|
Liao S, Amcoff M, Nässel DR. Impact of high-fat diet on lifespan, metabolism, fecundity and behavioral senescence in Drosophila. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 133:103495. [PMID: 33171202 DOI: 10.1016/j.ibmb.2020.103495] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/01/2020] [Accepted: 10/30/2020] [Indexed: 06/11/2023]
Abstract
Excess consumption of high-fat diet (HFD) is likely to result in obesity and increases the predisposition to associated health disorders. Drosophila melanogaster has emerged as an important model to study the effects of HFD on metabolism, gut function, behavior, and ageing. In this study, we investigated the effects of HFD on physiology and behavior of female flies at different time-points over several weeks. We found that HFD decreases lifespan, and also with age leads to accelerated decline of climbing ability in both virgins and mated flies. In virgins HFD also increased sleep fragmentation with age. Furthermore, long-term exposure to HFD results in elevated adipokinetic hormone (AKH) transcript levels and an enlarged crop with increased lipid stores. We detected no long-term effects of HFD on body mass, or levels of triacylglycerides (TAG), glycogen or glucose, although fecundity was diminished. However, one week of HFD resulted in decreased body mass and elevated TAG levels in mated flies. Finally, we investigated the role of AKH in regulating effects of HFD during aging. Both with normal diet (ND) and HFD, Akh mutant flies displayed increased longevity compared to control flies. However, both mutants and controls showed shortened lifespan on HFD compared to ND. In flies exposed to ND, fecundity is decreased in Akh mutants compared to controls after one week, but increased after three weeks. However, HFD leads to a similar decrease in fecundity in both genotypes after both exposure times. Thus, long-term exposure to HFD increases AKH signaling, impairs lifespan and fecundity and augments age-related behavioral senescence.
Collapse
Affiliation(s)
- Sifang Liao
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Mirjam Amcoff
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
89
|
Baldridge D, Wangler MF, Bowman AN, Yamamoto S, Schedl T, Pak SC, Postlethwait JH, Shin J, Solnica-Krezel L, Bellen HJ, Westerfield M. Model organisms contribute to diagnosis and discovery in the undiagnosed diseases network: current state and a future vision. Orphanet J Rare Dis 2021; 16:206. [PMID: 33962631 PMCID: PMC8103593 DOI: 10.1186/s13023-021-01839-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Decreased sequencing costs have led to an explosion of genetic and genomic data. These data have revealed thousands of candidate human disease variants. Establishing which variants cause phenotypes and diseases, however, has remained challenging. Significant progress has been made, including advances by the National Institutes of Health (NIH)-funded Undiagnosed Diseases Network (UDN). However, 6000-13,000 additional disease genes remain to be identified. The continued discovery of rare diseases and their genetic underpinnings provides benefits to affected patients, of whom there are more than 400 million worldwide, and also advances understanding the mechanisms of more common diseases. Platforms employing model organisms enable discovery of novel gene-disease relationships, help establish variant pathogenicity, and often lead to the exploration of underlying mechanisms of pathophysiology that suggest new therapies. The Model Organism Screening Center (MOSC) of the UDN is a unique resource dedicated to utilizing informatics and functional studies in model organisms, including worm (Caenorhabditis elegans), fly (Drosophila melanogaster), and zebrafish (Danio rerio), to aid in diagnosis. The MOSC has directly contributed to the diagnosis of challenging cases, including multiple patients with complex, multi-organ phenotypes. In addition, the MOSC provides a framework for how basic scientists and clinicians can collaborate to drive diagnoses. Customized experimental plans take into account patient presentations, specific genes and variant(s), and appropriateness of each model organism for analysis. The MOSC also generates bioinformatic and experimental tools and reagents for the wider scientific community. Two elements of the MOSC that have been instrumental in its success are (1) multidisciplinary teams with expertise in variant bioinformatics and in human and model organism genetics, and (2) mechanisms for ongoing communication with clinical teams. Here we provide a position statement regarding the central role of model organisms for continued discovery of disease genes, and we advocate for the continuation and expansion of MOSC-type research entities as a Model Organisms Network (MON) to be funded through grant applications submitted to the NIH, family groups focused on specific rare diseases, other philanthropic organizations, industry partnerships, and other sources of support.
Collapse
Affiliation(s)
- Dustin Baldridge
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, 77030, USA.
- Department of Pediatrics, BCM, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA.
| | - Angela N Bowman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA
- Department of Neuroscience, BCM, Houston, TX, 77030, USA
| | - Tim Schedl
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Stephen C Pak
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Jimann Shin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX, 77030, USA
- Department of Neuroscience, BCM, Houston, TX, 77030, USA
- Howard Hughes Medical Institute, Houston, TX, 77030, USA
| | - Monte Westerfield
- Institute of Neuroscience, University of Oregon, Eugene, OR, 97403, USA
| |
Collapse
|
90
|
Choi BJ, Chen YCD, Desplan C. Building a circuit through correlated spontaneous neuronal activity in the developing vertebrate and invertebrate visual systems. Genes Dev 2021; 35:677-691. [PMID: 33888564 PMCID: PMC8091978 DOI: 10.1101/gad.348241.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
During the development of the vertebrate nervous systems, genetic programs assemble an immature circuit that is subsequently refined by neuronal activity evoked by external stimuli. However, prior to sensory experience, the intrinsic property of the developing nervous system also triggers correlated network-level neuronal activity, with retinal waves in the developing vertebrate retina being the best documented example. Spontaneous activity has also been found in the visual system of Drosophila Here, we compare the spontaneous activity of the developing visual system between mammalian and Drosophila and suggest that Drosophila is an emerging model for mechanistic and functional studies of correlated spontaneous activity.
Collapse
Affiliation(s)
- Ben Jiwon Choi
- Department of Biology, New York University, New York, New York 10003, USA
| | | | - Claude Desplan
- Department of Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
91
|
Derby CD. The Crustacean Antennule: A Complex Organ Adapted for Lifelong Function in Diverse Environments and Lifestyles. THE BIOLOGICAL BULLETIN 2021; 240:67-81. [PMID: 33939945 DOI: 10.1086/713537] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
AbstractThe crustacean first antenna, or antennule, has been an experimental model for studying sensory biology for over 150 years. Investigations have led to a clearer understanding of the functional organization of the antennule as an olfactory organ but also to a realization that the antennule is much more than that. Across the Crustacea, the antennules take on many forms and functions. As an example, the antennule of reptantian decapods has many types of sensilla, each with distinct structure and function and with hundreds of thousands of chemosensory neurons expressing hundreds of genes that code for diverse classes of receptor proteins. Together, these antennular sensilla represent multiple chemosensory pathways, each with its own central connections and functions. The antennule also has a diversity of sensors of mechanical stimuli, including vibrations, touch, water flow, and the animal's own movements. The antennule likely also detects other environmental cues, such as temperature, oxygen, pH, salinity, and noxious stimuli. Furthermore, the antennule is a motor organ-it is flicked to temporally and spatially sample the animal's chemo-mechanical surroundings-and this information is used in resolving the structure of chemical plumes and locating the odor source. The antennule is also adapted to maintain lifelong function in a changing environment. For example, it has specific secretory glands, grooming structures, and behaviors to stay clean and functional. Antennular sensilla and the annuli on which they reside are also added and replaced, leading to a complete turnover of the antennule over several molts. Thus, the antennule is a complex and dynamic sensory-motor integrator that is intricately engaged in most aspects of the lives of crustaceans.
Collapse
|
92
|
Nweke EE, Thimiri Govinda Raj DB. Development of insect cell line using CRISPR technology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 180:1-20. [PMID: 33934833 DOI: 10.1016/bs.pmbts.2021.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In this chapter, we delineated the methods of CRISPR technology that has been used for the development of engineered insect cell line. We elaborated on how CRISPR/Cas9 genome editing in Drosophila melanogaster, Bombyx mori, Spodoptera frugiperda (Sf9 and Sf21), and Mosquitoes enabled the use of model or non-model insect system in various biological and medical applications. Also, the application of synthetic baculovirus genome along with CRISPR/Cas9 vector system to enable genome editing of insect cell systems for treatment of communicable and non-communicable diseases.
Collapse
Affiliation(s)
| | - Deepak B Thimiri Govinda Raj
- Synthetic Nanobiotechnology and Biomachines Group, ERA Synthetic Biology, Centre for Synthetic Biology and Precision Medicine, CSIR, Pretoria, South Africa.
| |
Collapse
|
93
|
Wang X, Verschut TA, Billeter JC, Maan ME. Seven Questions on the Chemical Ecology and Neurogenetics of Resource-Mediated Speciation. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.640486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Adaptation to different environments can result in reproductive isolation between populations and the formation of new species. Food resources are among the most important environmental factors shaping local adaptation. The chemosensory system, the most ubiquitous sensory channel in the animal kingdom, not only detects food resources and their chemical composition, but also mediates sexual communication and reproductive isolation in many taxa. Chemosensory divergence may thus play a crucial role in resource-mediated adaptation and speciation. Understanding how the chemosensory system can facilitate resource-mediated ecological speciation requires integrating mechanistic studies of the chemosensory system with ecological studies, to link the genetics and physiology of chemosensory properties to divergent adaptation. In this review, we use examples of insect research to present seven key questions that can be used to understand how the chemosensory system can facilitate resource-mediated ecological speciation in consumer populations.
Collapse
|
94
|
Mishra P, Yang SE, Montgomery AB, Reed AR, Rodan AR, Rothenfluh A. The fly liquid-food electroshock assay (FLEA) suggests opposite roles for neuropeptide F in avoidance of bitterness and shock. BMC Biol 2021; 19:31. [PMID: 33593351 PMCID: PMC7888162 DOI: 10.1186/s12915-021-00969-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/29/2021] [Indexed: 12/03/2022] Open
Abstract
Background Proper regulation of feeding is important for an organism’s well-being and survival and involves a motivational component directing the search for food. Dissecting the molecular and neural mechanisms of motivated feeding behavior requires assays that allow quantification of both motivation and food intake. Measurements of motivated behavior usually involve assessing physical effort or overcoming an aversive stimulus. Food intake in Drosophila can be determined in a number of ways, including by measuring the time a fly’s proboscis interacts with a food source associated with an electrical current in the fly liquid-food interaction counter (FLIC). Here, we show that electrical current flowing through flies during this interaction is aversive, and we describe a modified assay to measure motivation in Drosophila. Results Food intake is reduced during the interaction with FLIC when the electrical current is turned on, which provides a confounding variable in studies of motivated behavior. Based on the FLIC, we engineer a novel assay, the fly liquid-food electroshock assay (FLEA), which allows for current adjustments for each feeding well. Using the FLEA, we show that both external incentives and internal motivational state can serve as drivers for flies to overcome higher current (electric shock) to obtain superior food. Unlike similar assays in which bitterness is the aversive stimulus for the fly to overcome, we show that current perception is not discounted as flies become more food-deprived. Finally, we use genetically manipulated flies to show that neuropeptide F, an orthologue of mammalian NPY previously implicated in regulation of feeding motivation, is required for sensory processing of electrical current. Conclusion The FLEA is therefore a novel assay to accurately measure incentive motivation in Drosophila. Using the FLEA, we also show that neuropeptide F is required for proper perception or processing of an electroshock, a novel function for this neuropeptide involved in the processing of external and internal stimuli. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-00969-7.
Collapse
Affiliation(s)
- Puskar Mishra
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA.,Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Shany E Yang
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | | | - Addison R Reed
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA.,Department of Internal Medicine, Division of Nephrology & Hypertension, University of Utah, Salt Lake City, UT, USA.,Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.,Medical Service, Veterans Affairs Salt Lake City Health Care System, University of Utah, Salt Lake City, UT, USA
| | - Adrian Rothenfluh
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA. .,Department of Human Genetics, University of Utah, Salt Lake City, UT, USA. .,Department of Psychiatry, University of Utah, Salt Lake City, UT, USA. .,Department of Neurobiology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
95
|
Ke M, Chong CM, Zhu Q, Zhang K, Cai CZ, Lu JH, Qin D, Su H. Comprehensive Perspectives on Experimental Models for Parkinson's Disease. Aging Dis 2021; 12:223-246. [PMID: 33532138 PMCID: PMC7801282 DOI: 10.14336/ad.2020.0331] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/31/2020] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) ranks second among the most common neurodegenerative diseases, characterized by progressive and selective loss of dopaminergic neurons. Various cross-species preclinical models, including cellular models and animal models, have been established through the decades to study the etiology and mechanism of the disease from cell lines to nonhuman primates. These models are aimed at developing effective therapeutic strategies for the disease. None of the current models can replicate all major pathological and clinical phenotypes of PD. Selection of the model for PD largely relies on our interest of study. In this review, we systemically summarized experimental PD models, including cellular and animal models used in preclinical studies, to understand the pathogenesis of PD. This review is intended to provide current knowledge about the application of these different PD models, with focus on their strengths and limitations with respect to their contributions to the assessment of the molecular pathobiology of PD and identification of the therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Minjing Ke
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cheong-Meng Chong
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qi Zhu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ke Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cui-Zan Cai
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jia-Hong Lu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dajiang Qin
- 2Guangzhou Regenerative Medicine and Health Guangdong Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,3South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Huanxing Su
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
96
|
Kohsaka H, Nose A. Optogenetics in Drosophila. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:309-320. [PMID: 33398822 DOI: 10.1007/978-981-15-8763-4_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The fruit fly Drosophila melanogaster, an insect 4 mm long, has served as the experimental subject in a wide range of biological research, including neuroscience. In this chapter, we briefly introduce optogenetic applications in Drosophila neuroscience research. First, we describe the development of Drosophila from egg to adult. In fly neuroscience, temperature-controlled perturbation of neural activity, sometimes called "thermogenetics," has been an invaluable tool that predates the advent of optogenetics. After briefly introducing this perturbation technique, we describe the process of generating transgenic flies that express optogenetic probes in a specific group of cells. Transgenic techniques are crucial in the application of optogenetics in Drosophila neuroscience; here we introduce the transposon P-elements, ϕC31 integrase, and CRISPR-Cas9 methods. As for cell-specific gene expression techniques, the binary expression systems utilizing Gal4-UAS, LexA-lexAop, and Q-system are described. We also present a short and basic optogenetic experiment with Drosophila larvae as a practical example. Finally, we review a few recent studies in Drosophila neuroscience that made use of optogenetics. In this overview of fly development, transgenic methods, and applications of optogenetics, we present an introductory background to optogenetics in Drosophila.
Collapse
Affiliation(s)
- Hiroshi Kohsaka
- Department of Complexity Science and Engineering, University of Tokyo, Kashiwanoha, Chiba, Japan.
| | - Akinao Nose
- Department of Complexity Science and Engineering, University of Tokyo, Kashiwanoha, Chiba, Japan.,Department of Physics, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
97
|
Ramos-Hryb AB, Ramirez MF, Lino-de-Oliveira C, Pagani MR. Stress-mediated hyperactivity and anhedonia resistant to diazepam and fluoxetine in drosophila. Stress 2021; 24:96-106. [PMID: 32319840 DOI: 10.1080/10253890.2020.1759547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Distresses may induce behavioral phenotypes constituting heuristic models for psychopharmacology studies. In several species, including Drosophila, antidepressants counteract stress-induced phenotypes allowing the use of these models to test new psychoactive drugs. Here, we developed a novel and time-efficient protocol to provoke stress-induced phenotypes in Drosophila for the study of psychopharmacological agents. In the first experiment, flies (n = 12/groups) were exposed to a random-sequence of different types of stresses during nearly 24 h (including social isolation, fasting, heat, and electric shock), a protocol named short-term variable stress (SVS). Second, flies were exposed to a single stressful stimulus (social isolation, fasting, heat shock or electric shock, n = 12/groups). Next, flies submitted to SVS protocol were treated with vehicle, diazepam or fluoxetine (n = 12/groups). At the end of the stress protocols, behavioral phenotypes were evaluated in the open field (OF) and sucrose preference tests. In comparison to the unstressed group, flies exposed to SVS exhibited hyperactivity, as well as shorter times exploring the boundaries of the OF. In contrast to fasting stress, SVS reduced sucrose preference in flies. By analyzing the effects of individual stimuli on fly behavior, fasting and electric shock appear to be the predominant influences on the SVS-induced behaviors. Although fluoxetine or diazepam reduced the initial locomotor activity of flies, no treatment prevented the sequelae of SVS. Altogether, this study provides a time-efficient model system for the study of stress-mediated hyperactivity and anhedonia-like state resistant to fluoxetine and diazepam. The applications of SVS in Drosophila to preclinical psychopharmacology require further studies. LAY SUMMARY Exposition to unpredictable stress plays a significant role in psychiatric disorder's onset. Behavioral traits of these disorders can be partially modeled in rodents aimed at developing psychopharmacological therapies. However, studies in rodents were questioned by ethical issues. Focused on 3Rs principles, we developed a preclinical model for stress and psychopharmacology research in Drosophila. Variable stress induced behavioral alterations, including hyperlocomotion and reduced preference for sucrose in flies. However, behavioral alterations were resistant to fluoxetine and diazepam.
Collapse
Affiliation(s)
- Ana Belén Ramos-Hryb
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
- Postgraduation Program in Pharmacology, CCB, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Mauro Federico Ramirez
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Cilene Lino-de-Oliveira
- Postgraduation Program in Pharmacology, CCB, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiological Sciences, CCB, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Mario Rafael Pagani
- Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|
98
|
Niccoli T, Kerr F, Snoeren I, Fabian D, Aleyakpo B, Ivanov D, Sofola-Adesakin O, Cryar A, Adcott J, Thornton J, Partridge L. Activating transcription factor 4-dependent lactate dehydrogenase activation as a protective response to amyloid beta toxicity. Brain Commun 2021; 3:fcab053. [PMID: 33977265 PMCID: PMC8093921 DOI: 10.1093/braincomms/fcab053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 02/07/2022] [Accepted: 02/09/2021] [Indexed: 12/21/2022] Open
Abstract
Accumulation of amyloid beta peptides is thought to initiate the pathogenesis of Alzheimer's disease. However, the precise mechanisms mediating their neurotoxicity are unclear. Our microarray analyses show that, in Drosophila models of amyloid beta 42 toxicity, genes involved in the unfolded protein response and metabolic processes are upregulated in brain. Comparison with the brain transcriptome of early-stage Alzheimer's patients revealed a common transcriptional signature, but with generally opposing directions of gene expression changes between flies and humans. Among these differentially regulated genes, lactate dehydrogenase (Ldh) was up-regulated by the greatest degree in amyloid beta 42 flies and the human orthologues (LDHA and LDHB) were down-regulated in patients. Functional analyses revealed that either over-expression or inhibition of Ldh by RNA interference (RNAi) slightly exacerbated climbing defects in both healthy and amyloid beta 42-induced Drosophila. This suggests that metabolic responses to lactate dehydrogenase must be finely-tuned, and that its observed upregulation following amyloid beta 42 production could potentially represent a compensatory protection to maintain pathway homeostasis in this model, with further manipulation leading to detrimental effects. The increased Ldh expression in amyloid beta 42 flies was regulated partially by unfolded protein response signalling, as ATF4 RNAi diminished the transcriptional response and enhanced amyloid beta 42-induced climbing phenotypes. Further functional studies are required to determine whether Ldh upregulation provides compensatory neuroprotection against amyloid beta 42-induced loss of activating transcription factor 4 activity and endoplasmatic reticulum stress. Our study thus reveals dysregulation of lactate dehydrogenase signalling in Drosophila models and patients with Alzheimer's disease, which may lead to a detrimental loss of metabolic homeostasis. Importantly, we observed that down-regulation of ATF4-dependent endoplasmic reticulum-stress signalling in this context appears to prevent Ldh compensation and to exacerbate amyloid beta 42-dependent neuronal toxicity. Our findings, therefore, suggest caution in the use of therapeutic strategies focussed on down-regulation of this pathway for the treatment of Alzheimer's disease, since its natural response to the toxic peptide may induce beneficial neuroprotective effects.
Collapse
Affiliation(s)
- Teresa Niccoli
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Correspondence may also be addressed to: Teresa Niccoli E-mail:
| | - Fiona Kerr
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Department of Biological and Biomedical Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK
- Department of Life Sciences, School of Applied Sciences, Edinburgh Napier University, Edinburgh EH11 4BN, UK
- Correspondence may also be addressed to: Fiona Kerr Department of Life Sciences, School of Applied Sciences, Edinburgh Napier University, Sighthill Campus Edinburgh EH11 4BN, UK E-mail:
| | - Inge Snoeren
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Daniel Fabian
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Benjamin Aleyakpo
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Dobril Ivanov
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
- UK Dementia Research Institute (UKDRI), Cardiff University, Cardiff CF24 4HQ, UK
| | - Oyinkan Sofola-Adesakin
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Adam Cryar
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Jennifer Adcott
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
| | - Janet Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London WC1E 6BT, UK
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
- Correspondence to: Linda Partridge Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London Darwin Building, Gower Street, London WC1E 6BT, UK E-mail:
| |
Collapse
|
99
|
Chvilicek MM, Titos I, Rothenfluh A. The Neurotransmitters Involved in Drosophila Alcohol-Induced Behaviors. Front Behav Neurosci 2020; 14:607700. [PMID: 33384590 PMCID: PMC7770116 DOI: 10.3389/fnbeh.2020.607700] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alcohol is a widely used and abused substance with numerous negative consequences for human health and safety. Historically, alcohol's widespread, non-specific neurobiological effects have made it a challenge to study in humans. Therefore, model organisms are a critical tool for unraveling the mechanisms of alcohol action and subsequent effects on behavior. Drosophila melanogaster is genetically tractable and displays a vast behavioral repertoire, making it a particularly good candidate for examining the neurobiology of alcohol responses. In addition to being experimentally amenable, Drosophila have high face and mechanistic validity: their alcohol-related behaviors are remarkably consistent with humans and other mammalian species, and they share numerous conserved neurotransmitters and signaling pathways. Flies have a long history in alcohol research, which has been enhanced in recent years by the development of tools that allow for manipulating individual Drosophila neurotransmitters. Through advancements such as the GAL4/UAS system and CRISPR/Cas9 mutagenesis, investigation of specific neurotransmitters in small subsets of neurons has become ever more achievable. In this review, we describe recent progress in understanding the contribution of seven neurotransmitters to fly behavior, focusing on their roles in alcohol response: dopamine, octopamine, tyramine, serotonin, glutamate, GABA, and acetylcholine. We chose these small-molecule neurotransmitters due to their conservation in mammals and their importance for behavior. While neurotransmitters like dopamine and octopamine have received significant research emphasis regarding their contributions to behavior, others, like glutamate, GABA, and acetylcholine, remain relatively unexplored. Here, we summarize recent genetic and behavioral findings concerning these seven neurotransmitters and their roles in the behavioral response to alcohol, highlighting the fitness of the fly as a model for human alcohol use.
Collapse
Affiliation(s)
- Maggie M. Chvilicek
- Department of Psychiatry, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT, United States
| | - Iris Titos
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| | - Adrian Rothenfluh
- Department of Psychiatry, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
- Neuroscience Graduate Program, University of Utah, Salt Lake City, UT, United States
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, United States
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
100
|
Inducing different severities of traumatic brain injury in Drosophila using a piezoelectric actuator. Nat Protoc 2020; 16:263-282. [PMID: 33277631 DOI: 10.1038/s41596-020-00415-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022]
Abstract
Drosophila models have been instrumental in providing insights into molecular mechanisms of neurodegeneration, with wide application to human disease. The brain degeneration associated with traumatic brain injury (TBI) has been modeled in Drosophila using devices that inflict trauma on multiple parts of the fly body, including the head. However, the injuries produced by these models are not specific in location and are inconsistent between individual animals. We have recently developed a device that can be used to inflict controlled head injury to flies, resulting in physiological responses that are remarkably similar to those observed in humans with TBI. This protocol describes the construction, calibration and use of the Drosophila TBI (dTBI) device, a platform that employs a piezoelectric actuator to reproducibly deliver a force in order to briefly compress the fly head against a metal surface. The extent of head compression can be controlled through an electrical circuit, allowing the operator to set different levels of injury. The entire device can be assembled and calibrated in under a week. The device components and the necessary electrical tools are readily available and cost ~$800. The dTBI device can be used to harness the power of Drosophila genetics and perform large-scale genetic or pharmacological screens, using a 7-d post-injury survival curve to identify modifiers of injury.
Collapse
|