1
|
Schloss SS, Marshall ZQ, Santistevan NJ, Gjorcheska S, Stenzel A, Barske L, Nelson JC. Cadherin-16 regulates acoustic sensory gating in zebrafish through endocrine signaling. PLoS Biol 2025; 23:e3003164. [PMID: 40315416 PMCID: PMC12077787 DOI: 10.1371/journal.pbio.3003164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 05/14/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025] Open
Abstract
Sensory thresholds enable animals to regulate their behavioral responses to environmental threats. Despite the importance of sensory thresholds for animal behavior and human health, we do not yet have a full appreciation of the underlying molecular-genetic and circuit mechanisms. The larval zebrafish acoustic startle response provides a powerful system to identify molecular mechanisms underlying establishment of sensory thresholds and plasticity of thresholds through mechanisms like habituation. Using this system, we identify Cadherin-16 as a previously undescribed regulator of sensory gating. We demonstrate that Cadherin-16 regulates sensory thresholds via an endocrine organ, the corpuscle of Stannius (CS), which is essential in zebrafish for regulating Ca2+ homeostasis. We further show that Cadherin-16 regulates whole-body calcium and ultimately behavior through the hormone Stanniocalcin 1l (Stc1l), and the IGF-regulatory metalloprotease, Papp-aa. Finally, we demonstrate the importance of the CS through ablation experiments that reveal its role in promoting normal acoustic sensory gating. Together, our results uncover a previously undescribed brain non-autonomous pathway for the regulation of behavior and underscore Ca2+ homeostasis as a critical process underlying sensory gating in vivo.
Collapse
Affiliation(s)
- Susannah S. Schloss
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, United States of America
| | - Zackary Q. Marshall
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, United States of America
| | - Nicholas J. Santistevan
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, United States of America
| | - Stefani Gjorcheska
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Amanda Stenzel
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, United States of America
| | - Lindsey Barske
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
2
|
Stevanović M, Tagkalidou N, Multisanti CR, Pujol S, Aljabasini O, Prats E, Faggio C, Porta JM, Barata C, Raldúa D. Zebra_K, a kinematic analysis automated platform for assessing sensitivity, habituation and prepulse inhibition of the acoustic startle response in adult zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:178028. [PMID: 39681027 DOI: 10.1016/j.scitotenv.2024.178028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/18/2024] [Accepted: 12/07/2024] [Indexed: 12/18/2024]
Abstract
The acoustic startle response (ASR) is leaded by a sudden and intense acoustic stimulus. ASR has several forms of plasticity, including habituation and sensorimotor gating. Although ASR and its plasticity have been intensively studied in zebrafish (Danio rerio) larvae, information in adult zebrafish is still very scarce. In this manuscript we present Zebra_K, a new automated high-content kinematic analysis platform for assessing ASR, its habituation and prepulse inhibition (PPI), a quantitative measure of sensorimotor gating, in adult zebrafish. The analysis of the kinematic parameters of ASR in adult zebrafish has shown a single response wave consistent with the short-latency C-bend described in zebrafish larvae. Moreover, protocols have been designed and validated in Zebra_K for the analysis of sensitivity, habituation and PPI of this response. Then, the effect of the time of day and the gender on zebrafish ASR plasticity has been analyzed for the first time. Females exhibited higher responsiveness and a lower habituation and PPI than males, a result consistent with the gender effect described in other animal models and in humans. This platform has also been used to determine the effect of a pharmacological modulators of ASR plasticity, the NMDA-receptor antagonist ketamine. As described in other animal models, ketamine increased the responsiveness to the acoustic stimuli, decreasing habituation and leading to complete abolition of PPI. These results enhance the interest of using adult zebrafish to assess the potential effect of environmental pollutants on ASR plasticity.
Collapse
Affiliation(s)
- Marija Stevanović
- Institute of Pesticides and Environmental Protection, Banatska 31b, 11080 Belgrade, Serbia
| | - Niki Tagkalidou
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona, 18, 08034 Barcelona, Spain
| | - Cristiana Roberta Multisanti
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci snc, 98168 Messina, Italy; Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy
| | - Sergi Pujol
- Institut de Robòtica i Informàtica Industrial, CSIC-UPC, Barcelona, Spain
| | - Ouwais Aljabasini
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona, 18, 08034 Barcelona, Spain
| | - Eva Prats
- Research and Development Center (CID-CSIC), Jordi Girona, 18, 08034 Barcelona, Spain
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres, 31, 98166 Messina, Italy; Department of Eco-Sustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, 80122 Naples, Italy
| | - Josep Maria Porta
- Institut de Robòtica i Informàtica Industrial, CSIC-UPC, Barcelona, Spain
| | - Carlos Barata
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona, 18, 08034 Barcelona, Spain
| | - Demetrio Raldúa
- Institute for Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona, 18, 08034 Barcelona, Spain.
| |
Collapse
|
3
|
Dya GA, Lebedeva OS, Gushchevarov DA, Volovikov EA, Belikova LD, Kopylova IV, Postnikov AB, Artemieva MM, Medvedeva NA, Lagarkova MA, Katrukha AG, Serebryanaya DV. Specific cleavage of IGFBP-4 by papp-a in nervous tissue. Biochem Biophys Res Commun 2024; 733:150655. [PMID: 39244846 DOI: 10.1016/j.bbrc.2024.150655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Astrocytes are subtypes of glial cells involved in metabolic, structural, homeostatic, and neuroprotective processes that help neurons maintain viability. Insulin-like growth factors IGF-1 and IGF-2 are known to have neuroprotective effects on neurons and glial cells through interaction with specific receptors. IGF forms a complex with IGF-binding proteins (IGFBP) in nervous tissue and is released from the complex via IGFBP proteolysis by specific proteases. It has been reported that IGFBP-2, 5 and 6 are cleaved by specific proteases in the central nervous system (CNS), followed by IGF release; however, it was unknown whether IGFBP-4 was exposed to a particular proteolysis in nervous tissue. Using neurons and astrocytes derived from human induced pluripotent stem cell lines (hiPSC), as well as rat brain-sourced primary neuron-glia cultures, we demonstrated that IGFBP-4 is specifically cleaved in nervous tissue by the Pregnancy Associated Plasma Protein A (PAPP-A) protease and that this cleavage is IGF-dependent. Our results indicate that astrocyte rather than neuron PAPP-A cleaves IGFBP-4 in nervous tissue suggesting that this may be one of the fundamental mechanisms for IGF interchange between these two types of cells.
Collapse
Affiliation(s)
- German A Dya
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga S Lebedeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | | | - Egor A Volovikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Lilia D Belikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Irina V Kopylova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | | | | | | | - Maria A Lagarkova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia; Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Alexey G Katrukha
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia; Hytest, Turku, Finland
| | - Daria V Serebryanaya
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia; Pirogov Russian National Research Medical University, Moscow, Russia.
| |
Collapse
|
4
|
Santistevan NJ, Ford CT, Gilsdorf CS, Grinblat Y. Behavioral and transcriptomic analyses of mecp2 function in zebrafish. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32981. [PMID: 38551133 DOI: 10.1002/ajmg.b.32981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 11/15/2024]
Abstract
Rett syndrome (RTT), a human neurodevelopmental disorder characterized by severe cognitive and motor impairments, is caused by dysfunction of the conserved transcriptional regulator Methyl-CpG-binding protein 2 (MECP2). Genetic analyses in mouse Mecp2 mutants, which exhibit key features of human RTT, have been essential for deciphering the mechanisms of MeCP2 function; nonetheless, our understanding of these complex mechanisms is incomplete. Zebrafish mecp2 mutants exhibit mild behavioral deficits but have not been analyzed in depth. Here, we combine transcriptomic and behavioral assays to assess baseline and stimulus-evoked motor responses and sensory filtering in zebrafish mecp2 mutants from 5 to 7 days post-fertilization (dpf). We show that zebrafish mecp2 function is required for normal thigmotaxis but is dispensable for gross movement, acoustic startle response, and sensory filtering (habituation and sensorimotor gating), and reveal a previously unknown role for mecp2 in behavioral responses to visual stimuli. RNA-seq analysis identified a large gene set that requires mecp2 function for correct transcription at 4 dpf, and pathway analysis revealed several pathways that require MeCP2 function in both zebrafish and mammals. These findings show that MeCP2's function as a transcriptional regulator is conserved across vertebrates and supports using zebrafish to complement mouse modeling in elucidating these conserved mechanisms.
Collapse
Affiliation(s)
- Nicholas J Santistevan
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | - Colby T Ford
- School of Data Science, University of North Carolina, Charlotte, North Carolina, USA
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, North Carolina, USA
- Tuple, LLC, Charlotte, North Carolina, USA
| | - Cole S Gilsdorf
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
| | - Yevgenya Grinblat
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Schloss SS, Marshall ZQ, Santistevan NJ, Gjorcheska S, Stenzel A, Barske L, Nelson JC. Cadherin 16 promotes sensory gating via the endocrine corpuscles of Stannius. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614609. [PMID: 39386705 PMCID: PMC11463452 DOI: 10.1101/2024.09.23.614609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Sensory thresholds enable animals to regulate their behavioral responses to environmental threats. Despite the importance of sensory thresholds for animal behavior and human health, we do not yet have a full appreciation of the underlying molecular-genetic and circuit mechanisms. The larval zebrafish acoustic startle response provides a powerful system to identify molecular mechanisms underlying establishment of sensory thresholds and plasticity of thresholds through mechanisms like habituation. Using this system, we identify Cadherin 16 as a previously undescribed regulator of sensory gating. We demonstrate that Cadherin 16 regulates sensory thresholds via an endocrine organ, the corpuscle of Stannius (CS), which is essential in zebrafish for regulating Ca2+ homeostasis. We further show that Cadherin 16 regulates whole-body calcium and ultimately behavior through the hormone Stanniocalcin 1L, and the IGF-regulatory metalloprotease, Papp-aa. Finally, we demonstrate the importance of the CS through ablation experiments that reveal its role in promoting normal acoustic sensory gating. Together, our results uncover a previously undescribed brain non-autonomous pathway for the regulation of behavior and establish Ca2+ homeostasis as a critical process underlying sensory gating in vivo.
Collapse
Affiliation(s)
- Susannah S. Schloss
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Zackary Q. Marshall
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Nicholas J. Santistevan
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Stefani Gjorcheska
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Amanda Stenzel
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Lindsey Barske
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| |
Collapse
|
6
|
Sumathipala SH, Khan S, Kozol RA, Araki Y, Syed S, Huganir RL, Dallman JE. Context-dependent hyperactivity in syngap1a and syngap1b zebrafish models of SYNGAP1-related disorder. Front Mol Neurosci 2024; 17:1401746. [PMID: 39050824 PMCID: PMC11266194 DOI: 10.3389/fnmol.2024.1401746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/04/2024] [Indexed: 07/27/2024] Open
Abstract
Background and aims SYNGAP1-related disorder (SYNGAP1-RD) is a prevalent genetic form of Autism Spectrum Disorder and Intellectual Disability (ASD/ID) and is caused by de novo or inherited mutations in one copy of the SYNGAP1 gene. In addition to ASD/ID, SYNGAP1 disorder is associated with comorbid symptoms including treatment-resistant-epilepsy, sleep disturbances, and gastrointestinal distress. Mechanistic links between these diverse symptoms and SYNGAP1 variants remain obscure, therefore, our goal was to generate a zebrafish model in which this range of symptoms can be studied. Methods We used CRISPR/Cas9 to introduce frameshift mutations in the syngap1a and syngap1b zebrafish duplicates (syngap1ab) and validated these stable models for Syngap1 loss-of-function. Because SYNGAP1 is extensively spliced, we mapped splice variants to the two zebrafish syngap1a and b genes and identified mammalian-like isoforms. We then quantified locomotory behaviors in zebrafish syngap1ab larvae under three conditions that normally evoke different arousal states in wild-type larvae: aversive, high-arousal acoustic, medium-arousal dark, and low-arousal light stimuli. Results We show that CRISPR/Cas9 indels in zebrafish syngap1a and syngap1b produced loss-of-function alleles at RNA and protein levels. Our analyses of zebrafish Syngap1 isoforms showed that, as in mammals, zebrafish Syngap1 N- and C-termini are extensively spliced. We identified a zebrafish syngap1 α1-like variant that maps exclusively to the syngap1b gene. Quantifying locomotor behaviors showed that syngap1ab mutant larvae are hyperactive compared to wild-type but to differing degrees depending on the stimulus. Hyperactivity was most pronounced in low arousal settings, and hyperactivity was proportional to the number of mutant syngap1 alleles. Limitations Syngap1 loss-of-function mutations produce relatively subtle phenotypes in zebrafish compared to mammals. For example, while mouse Syngap1 homozygotes die at birth, zebrafish syngap1ab-/- survive to adulthood and are fertile, thus some aspects of symptoms in people with SYNGAP1-Related Disorder are not likely to be reflected in zebrafish. Conclusion Our data support mutations in zebrafish syngap1ab as causal for hyperactivity associated with elevated arousal that is especially pronounced in low-arousal environments.
Collapse
Affiliation(s)
- Sureni H. Sumathipala
- Department of Biology, University of Miami, Coral Gables, FL, United States
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Suha Khan
- Department of Biology, University of Miami, Coral Gables, FL, United States
| | - Robert A. Kozol
- Department of Biology, University of Miami, Coral Gables, FL, United States
- Department of Biological Sciences, St. John’s University, Queens, NY, United States
| | - Yoichi Araki
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, United States
| | - Richard L. Huganir
- Department of Neuroscience and Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Julia E. Dallman
- Department of Biology, University of Miami, Coral Gables, FL, United States
| |
Collapse
|
7
|
Zúñiga Mouret R, Greenbaum JP, Doll HM, Brody EM, Iacobucci EL, Roland NC, Simamora RC, Ruiz I, Seymour R, Ludwick L, Krawitz JA, Groneberg AH, Marques JC, Laborde A, Rajan G, Del Bene F, Orger MB, Jain RA. The adaptor protein 2 (AP2) complex modulates habituation and behavioral selection across multiple pathways and time windows. iScience 2024; 27:109455. [PMID: 38550987 PMCID: PMC10973200 DOI: 10.1016/j.isci.2024.109455] [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: 03/29/2023] [Revised: 01/28/2024] [Accepted: 03/06/2024] [Indexed: 10/04/2024] Open
Abstract
Animals constantly integrate sensory information with prior experience to select behavioral responses appropriate to the current situation. Genetic factors supporting this behavioral flexibility are often disrupted in neuropsychiatric conditions, such as the autism-linked ap2s1 gene which supports acoustically evoked habituation learning. ap2s1 encodes an AP2 endocytosis adaptor complex subunit, although its behavioral mechanisms and importance have been unclear. Here, we show that multiple AP2 subunits regulate acoustically evoked behavior selection and habituation learning in zebrafish. Furthermore, ap2s1 biases escape behavior choice in sensory modality-specific manners, and broadly regulates action selection across sensory contexts. We demonstrate that the AP2 complex functions acutely in the nervous system to modulate acoustically evoked habituation, suggesting several spatially and/or temporally distinct mechanisms through which AP2 regulates escape behavior selection and performance. Altogether, we show the AP2 complex coordinates action selection across diverse contexts, providing a vertebrate model for ap2s1's role in human conditions including autism spectrum disorder.
Collapse
Affiliation(s)
- Rodrigo Zúñiga Mouret
- Department of Biology, Haverford College, Haverford, PA 19041, USA
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA
| | - Jordyn P. Greenbaum
- Department of Biology, Haverford College, Haverford, PA 19041, USA
- The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Hannah M. Doll
- Department of Biology, Haverford College, Haverford, PA 19041, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison WI 53705, USA
| | - Eliza M. Brody
- Department of Biology, Haverford College, Haverford, PA 19041, USA
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia PA 19104, USA
| | | | | | - Roy C. Simamora
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Ivan Ruiz
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Rory Seymour
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Leanne Ludwick
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Jacob A. Krawitz
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Antonia H. Groneberg
- Champalimaud Neuroscience Programme, Champalimaud Foundation, 1400-038 Lisboa, Portugal
| | - João C. Marques
- Champalimaud Neuroscience Programme, Champalimaud Foundation, 1400-038 Lisboa, Portugal
| | - Alexandre Laborde
- Champalimaud Neuroscience Programme, Champalimaud Foundation, 1400-038 Lisboa, Portugal
| | - Gokul Rajan
- Sorbonne Université; INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Institut Curie, PSL Research University; INSERM U934, CNRS UMR3215, Paris, France
| | - Filippo Del Bene
- Sorbonne Université; INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Michael B. Orger
- Champalimaud Neuroscience Programme, Champalimaud Foundation, 1400-038 Lisboa, Portugal
| | - Roshan A. Jain
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| |
Collapse
|
8
|
Ortiz EA, Campbell PD, Nelson JC, Granato M. A single base pair substitution in zebrafish distinguishes between innate and acute startle behavior regulation. PLoS One 2024; 19:e0300529. [PMID: 38498506 PMCID: PMC10947677 DOI: 10.1371/journal.pone.0300529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Behavioral thresholds define the lowest stimulus intensities sufficient to elicit a behavioral response. Establishment of baseline behavioral thresholds during development is critical for proper responses throughout the animal's life. Despite the relevance of such innate thresholds, the molecular mechanisms critical to establishing behavioral thresholds during development are not well understood. The acoustic startle response is a conserved behavior whose threshold is established during development yet is subsequently acutely regulated. We have previously identified a zebrafish mutant line (escapist) that displays a decreased baseline or innate acoustic startle threshold. Here, we identify a single base pair substitution on Chromosome 25 located within the coding sequence of the synaptotagmin 7a (syt7a) gene that is tightly linked to the escapist acoustic hypersensitivity phenotype. By generating animals in which we deleted the syt7a open reading frame, and subsequent complementation testing with the escapist line, we demonstrate that loss of syt7a function is not the cause of the escapist behavioral phenotype. Nonetheless, escapist mutants provide a powerful tool to decipher the overlap between acute and developmental regulation of behavioral thresholds. Extensive behavioral analyses reveal that in escapist mutants the establishment of the innate acoustic startle threshold is impaired, while regulation of its acute threshold remains intact. Moreover, our behavioral analyses reveal a deficit in baseline responses to visual stimuli, but not in the acute regulation of responses to visual stimuli. Together, this work eliminates loss of syt7a as causative for the escapist phenotype and suggests that mechanisms that regulate the establishment of behavioral thresholds in escapist larvae can operate independently from those regulating acute threshold regulation.
Collapse
Affiliation(s)
- Elelbin A. Ortiz
- Department of Neuroscience, University of Pennsylvania, Pennsylvania, PA, United States of America
- Department of Cell and Developmental Biology, University of Pennsylvania, Pennsylvania, PA, United States of America
| | - Philip D. Campbell
- Department of Cell and Developmental Biology, University of Pennsylvania, Pennsylvania, PA, United States of America
- Department of Psychiatry, University of Pennsylvania, Pennsylvania, PA, United States of America
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania, Pennsylvania, PA, United States of America
| |
Collapse
|
9
|
Conover CA, Oxvig C. The Pregnancy-Associated Plasma Protein-A (PAPP-A) Story. Endocr Rev 2023; 44:1012-1028. [PMID: 37267421 DOI: 10.1210/endrev/bnad017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/01/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) was first identified in the early 1970s as a placental protein of unknown function, present at high concentrations in the circulation of pregnant women. In the mid-to-late 1990s, PAPP-A was discovered to be a metzincin metalloproteinase, expressed by many nonplacental cells, that regulates local insulin-like growth factor (IGF) activity through cleavage of high-affinity IGF binding proteins (IGFBPs), in particular IGFBP-4. With PAPP-A as a cell surface-associated enzyme, the reduced affinity of the cleavage fragments results in increased IGF available to bind and activate IGF receptors in the pericellular environment. This proteolytic regulation of IGF activity is important, since the IGFs promote proliferation, differentiation, migration, and survival in various normal and cancer cells. Thus, there has been a steady growth in investigation of PAPP-A structure and function outside of pregnancy. This review provides historical perspective on the discovery of PAPP-A and its structure and cellular function, highlights key studies of the first 50 years in PAPP-A research, and introduces new findings from recent years.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
10
|
Ortiz EA, Campbell PD, Nelson JC, Granato M. A single base pair substitution on Chromosome 25 in zebrafish distinguishes between development and acute regulation of behavioral thresholds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554673. [PMID: 37662318 PMCID: PMC10473726 DOI: 10.1101/2023.08.25.554673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Behavioral thresholds define the lowest stimulus intensities sufficient to elicit a behavioral response. Establishment of baseline behavioral thresholds during development is critical for proper responses throughout the animal's life. Despite the relevance of such innate thresholds, the molecular mechanisms critical to establishing behavioral thresholds during development are not well understood. The acoustic startle response is a conserved behavior whose threshold is established during development yet is subsequently acutely regulated. We have previously identified a zebrafish mutant line ( escapist ) that displays a decreased baseline or innate acoustic startle threshold. Here, we identify a single base pair substitution on Chromosome 25 located within the coding sequence of the synaptotagmin 7a ( syt7a ) gene that is tightly linked to the escapist acoustic hypersensitivity phenotype. By generating animals in which we deleted the syt7a open reading frame, and subsequent complementation testing with the escapist line, we demonstrate that loss of syt7a function is not the cause of the escapist behavioral phenotype. Nonetheless, escapist mutants provide a powerful tool to decipher the overlap between acute and developmental regulation of behavioral thresholds. Extensive behavioral analyses reveal that in escapist mutants the establishment of the innate acoustic startle threshold is impaired, while regulation of its acute threshold remains intact. Moreover, our behavioral analyses reveal a deficit in baseline responses to visual stimuli, but not in the acute regulation of responses to visual stimuli. Together, this work eliminates loss of syt7a as causative for the escapist phenotype and suggests that mechanisms that regulate the establishment of behavioral thresholds in escapist larvae can operate largely independently from those regulating acute threshold regulation.
Collapse
|
11
|
AlAbdi L, Desbois M, Rusnac DV, Sulaiman RA, Rosenfeld JA, Lalani S, Murdock DR, Burrage LC, Undiagnosed Diseases Network, Billie Au PY, Towner S, Wilson WG, Wong L, Brunet T, Strobl-Wildemann G, Burton JE, Hoganson G, McWalter K, Begtrup A, Zarate YA, Christensen EL, Opperman KJ, Giles AC, Helaby R, Kania A, Zheng N, Grill B, Alkuraya FS. Loss-of-function variants in MYCBP2 cause neurobehavioural phenotypes and corpus callosum defects. Brain 2023; 146:1373-1387. [PMID: 36200388 PMCID: PMC10319777 DOI: 10.1093/brain/awac364] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022] Open
Abstract
The corpus callosum is a bundle of axon fibres that connects the two hemispheres of the brain. Neurodevelopmental disorders that feature dysgenesis of the corpus callosum as a core phenotype offer a valuable window into pathology derived from abnormal axon development. Here, we describe a cohort of eight patients with a neurodevelopmental disorder characterized by a range of deficits including corpus callosum abnormalities, developmental delay, intellectual disability, epilepsy and autistic features. Each patient harboured a distinct de novo variant in MYCBP2, a gene encoding an atypical really interesting new gene (RING) ubiquitin ligase and signalling hub with evolutionarily conserved functions in axon development. We used CRISPR/Cas9 gene editing to introduce disease-associated variants into conserved residues in the Caenorhabditis elegans MYCBP2 orthologue, RPM-1, and evaluated functional outcomes in vivo. Consistent with variable phenotypes in patients with MYCBP2 variants, C. elegans carrying the corresponding human mutations in rpm-1 displayed axonal and behavioural abnormalities including altered habituation. Furthermore, abnormal axonal accumulation of the autophagy marker LGG-1/LC3 occurred in variants that affect RPM-1 ubiquitin ligase activity. Functional genetic outcomes from anatomical, cell biological and behavioural readouts indicate that MYCBP2 variants are likely to result in loss of function. Collectively, our results from multiple human patients and CRISPR gene editing with an in vivo animal model support a direct link between MYCBP2 and a human neurodevelopmental spectrum disorder that we term, MYCBP2-related developmental delay with corpus callosum defects (MDCD).
Collapse
Affiliation(s)
- Lama AlAbdi
- Department of Zoology, College of Science, King Saud University, Riyadh 11362, Saudi Arabia
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Muriel Desbois
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Domniţa-Valeria Rusnac
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Raashda A Sulaiman
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seema Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - David R Murdock
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Ping Yee Billie Au
- Department of Medical Genetics, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shelley Towner
- Pediatric Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - William G Wilson
- Pediatric Genetics, University of Virginia, Charlottesville, VA 22903, USA
| | - Lawrence Wong
- Department of Genetics, Northern California Kaiser Permanente, Oakland, CA 94611, USA
| | - Theresa Brunet
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Institute of Neurogenomics (ING), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Jennifer E Burton
- Department of Genetics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - George Hoganson
- Department of Genetics, University of Illinois College of Medicine at Peoria, Peoria, IL 61605, USA
| | - Kirsty McWalter
- Genedx, Inc., 207 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amber Begtrup
- Genedx, Inc., 207 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Yuri A Zarate
- Section of Genetics and Metabolism, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Elyse L Christensen
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Karla J Opperman
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
| | - Andrew C Giles
- Division of Medical Sciences, University of Northern British Columbia, Prince George, BC V2N 4Z9, Canada
| | - Rana Helaby
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Artur Kania
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC H3A 2B4, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC H3A 2B2, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Ning Zheng
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA 98101, USA
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| |
Collapse
|
12
|
Nelson JC, Shoenhard H, Granato M. Integration of cooperative and opposing molecular programs drives learning-associated behavioral plasticity. PLoS Genet 2023; 19:e1010650. [PMID: 36972301 PMCID: PMC10079226 DOI: 10.1371/journal.pgen.1010650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 04/06/2023] [Accepted: 02/02/2023] [Indexed: 03/29/2023] Open
Abstract
Habituation is a foundational learning process critical for animals to adapt their behavior to changes in their sensory environment. Although habituation is considered a simple form of learning, the identification of a multitude of molecular pathways including several neurotransmitter systems that regulate this process suggests an unexpected level of complexity. How the vertebrate brain integrates these various pathways to accomplish habituation learning, whether they act independently or intersect with one another, and whether they act via divergent or overlapping neural circuits has remained unclear. To address these questions, we combined pharmacogenetic pathway analysis with unbiased whole-brain activity mapping using the larval zebrafish. Based on our findings, we propose five distinct molecular modules for the regulation of habituation learning and identify a set of molecularly defined brain regions associated with four of the five modules. Moreover, we find that in module 1 the palmitoyltransferase Hip14 cooperates with dopamine and NMDA signaling to drive habituation, while in module 3 the adaptor protein complex subunit Ap2s1 drives habituation by antagonizing dopamine signaling, revealing two distinct and opposing roles for dopaminergic neuromodulation in the regulation of behavioral plasticity. Combined, our results define a core set of distinct modules that we propose act in concert to regulate habituation-associated plasticity, and provide compelling evidence that even seemingly simple learning behaviors in a compact vertebrate brain are regulated by a complex and overlapping set of molecular mechanisms.
Collapse
Affiliation(s)
- Jessica C. Nelson
- Department of Cell and Developmental Biology; University of Pennsylvania, Perelman School of Medicine; Philadelphia, Pennsylvania, United States of America
- Department of Cell and Developmental Biology; University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Hannah Shoenhard
- Department of Cell and Developmental Biology; University of Pennsylvania, Perelman School of Medicine; Philadelphia, Pennsylvania, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology; University of Pennsylvania, Perelman School of Medicine; Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
13
|
Mrinalini R, Tamilanban T, Naveen Kumar V, Manasa K. Zebrafish - The Neurobehavioural Model in Trend. Neuroscience 2022; 520:95-118. [PMID: 36549602 DOI: 10.1016/j.neuroscience.2022.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Zebrafish (Danio rerio) is currently in vogue as a prevalently used experimental model for studies concerning neurobehavioural disorders and associated fields. Since the 1960s, this model has succeeded in breaking most barriers faced in the hunt for an experimental model. From its appearance to its high parity with human beings genetically, this model renders itself as an advantageous experimental lab animal. Neurobehavioural disorders have always posed an arduous task in terms of their detection as well as in determining their exact etiology. They are still, in most cases, diseases of interest for inventing or discovering novel pharmacological interventions. Thus, the need for a harbinger experimental model for studying neurobehaviours is escalating. Ensuring the same model is used for studying several neuro-studies conserves the results from inter-species variations. For this, we need a model that satisfies all the pre-requisite conditions to be made the final choice of model for neurobehavioural studies. This review recapitulates the progress of zebrafish as an experimental model with its most up-to-the-minute advances in the area. Various tests, assays, and responses employed using zebrafish in screening neuroactive drugs have been tabulated effectively. The tools, techniques, protocols, and apparatuses that bolster zebrafish studies are discussed. The probable research that can be done using zebrafish has also been briefly outlined. The various breeding and maintenance methods employed, along with the information on various strains available and most commonly used, are also elaborated upon, supplementing Zebrafish's use in neuroscience.
Collapse
Affiliation(s)
- R Mrinalini
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203
| | - T Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203
| | - V Naveen Kumar
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203.
| | - K Manasa
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203
| |
Collapse
|
14
|
Shoenhard H, Jain RA, Granato M. The calcium-sensing receptor (CaSR) regulates zebrafish sensorimotor decision making via a genetically defined cluster of hindbrain neurons. Cell Rep 2022; 41:111790. [PMID: 36476852 PMCID: PMC9813870 DOI: 10.1016/j.celrep.2022.111790] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Decision making is a fundamental nervous system function that ranges widely in complexity and speed of execution. We previously established larval zebrafish as a model for sensorimotor decision making and identified the G-protein-coupled calcium-sensing receptor (CaSR) to be critical for this process. Here, we report that CaSR functions in neurons to dynamically regulate the bias between two behavioral outcomes: escapes and reorientations. By employing a computational guided transgenic strategy, we identify a genetically defined neuronal cluster in the hindbrain as a key candidate site for CaSR function. Finally, we demonstrate that transgenic CaSR expression targeting this cluster consisting of a few hundred neurons shifts behavioral bias in wild-type animals and restores decision making deficits in CaSR mutants. Combined, our data provide a rare example of a G-protein-coupled receptor that biases vertebrate sensorimotor decision making via a defined neuronal cluster.
Collapse
Affiliation(s)
- Hannah Shoenhard
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roshan A Jain
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Blok LER, Boon M, van Reijmersdal B, Höffler KD, Fenckova M, Schenck A. Genetics, molecular control and clinical relevance of habituation learning. Neurosci Biobehav Rev 2022; 143:104883. [PMID: 36152842 DOI: 10.1016/j.neubiorev.2022.104883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
Habituation is the most fundamental form of learning. As a firewall that protects our brain from sensory overload, it is indispensable for cognitive processes. Studies in humans and animal models provide increasing evidence that habituation is affected in autism and related monogenic neurodevelopmental disorders (NDDs). An integrated application of habituation assessment in NDDs and their animal models has unexploited potential for neuroscience and medical care. With the aim to gain mechanistic insights, we systematically retrieved genes that have been demonstrated in the literature to underlie habituation. We identified 258 evolutionarily conserved genes across species, describe the biological processes they converge on, and highlight regulatory pathways and drugs that may alleviate habituation deficits. We also summarize current habituation paradigms and extract the most decisive arguments that support the crucial role of habituation for cognition in health and disease. We conclude that habituation is a conserved, quantitative, cognition- and disease-relevant process that can connect preclinical and clinical work, and hence is a powerful tool to advance research, diagnostics, and treatment of NDDs.
Collapse
Affiliation(s)
- Laura Elisabeth Rosalie Blok
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Marina Boon
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Kira Daniela Höffler
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands; Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| |
Collapse
|
16
|
Santistevan NJ, Nelson JC, Ortiz EA, Miller AH, Kenj Halabi D, Sippl ZA, Granato M, Grinblat Y. cacna2d3, a voltage-gated calcium channel subunit, functions in vertebrate habituation learning and the startle sensitivity threshold. PLoS One 2022; 17:e0270903. [PMID: 35834485 PMCID: PMC9282658 DOI: 10.1371/journal.pone.0270903] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/18/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The ability to filter sensory information into relevant versus irrelevant stimuli is a fundamental, conserved property of the central nervous system and is accomplished in part through habituation learning. Synaptic plasticity that underlies habituation learning has been described at the cellular level, yet the genetic regulators of this plasticity remain poorly understood, as do circuits that mediate sensory filtering. METHODS To identify genes critical for plasticity, a forward genetic screen for zebrafish genes that mediate habituation learning was performed, which identified a mutant allele, doryp177, that caused reduced habituation of the acoustic startle response. In this study, we combine whole-genome sequencing with behavioral analyses to characterize and identify the gene affected in doryp177 mutants. RESULTS Whole-genome sequencing identified the calcium voltage-gated channel auxiliary subunit alpha-2/delta-3 (cacna2d3) as a candidate gene affected in doryp177 mutants. Behavioral characterization of larvae homozygous for two additional, independently derived mutant alleles of cacna2d3, together with failure of these alleles to complement doryp177, confirmed a critical role for cacna2d3 in habituation learning. Notably, detailed analyses of the acoustic response in mutant larvae also revealed increased startle sensitivity to acoustic stimuli, suggesting a broader role for cacna2d3 in controlling innate response thresholds to acoustic stimuli. CONCLUSIONS Taken together, our data demonstrate a critical role for cacna2d3 in sensory filtering, a process that is disrupted in human CNS disorders, e.g. ADHD, schizophrenia, and autism.
Collapse
Affiliation(s)
- Nicholas J. Santistevan
- Department of Integrative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, United States of America
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Elelbin A. Ortiz
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States of America
- Neuroscience Graduate Program, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Andrew H. Miller
- Department of Integrative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States of America
- Neuroscience Ph.D. Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Dima Kenj Halabi
- Department of Integrative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Zoë A. Sippl
- Department of Integrative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail: (MG); (YG)
| | - Yevgenya Grinblat
- Department of Integrative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin, United States of America
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail: (MG); (YG)
| |
Collapse
|
17
|
Yoon B, Yeung P, Santistevan N, Bluhm LE, Kawasaki K, Kueper J, Dubielzig R, VanOudenhove J, Cotney J, Liao EC, Grinblat Y. Zebrafish models of alx-linked frontonasal dysplasia reveal a role for Alx1 and Alx3 in the anterior segment and vasculature of the developing eye. Biol Open 2022; 11:bio059189. [PMID: 35142342 PMCID: PMC9167625 DOI: 10.1242/bio.059189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/28/2022] [Indexed: 11/18/2022] Open
Abstract
The cellular and genetic mechanisms that coordinate formation of facial sensory structures with surrounding skeletal and soft tissue elements remain poorly understood. Alx1, a homeobox transcription factor, is a key regulator of midfacial morphogenesis. ALX1 mutations in humans are linked to severe congenital anomalies of the facial skeleton (frontonasal dysplasia, FND) with malformation or absence of eyes and orbital contents (micro- and anophthalmia). Zebrafish with loss-of-function alx1 mutations develop with craniofacial and ocular defects of variable penetrance, likely due to compensatory upregulation in expression of a paralogous gene, alx3. Here we show that zebrafish alx1;alx3 mutants develop with highly penetrant cranial and ocular defects that resemble human ALX1-linked FND. alx1 and alx3 are expressed in anterior cranial neural crest (aCNC), which gives rise to the anterior neurocranium (ANC), anterior segment structures of the eye and vascular pericytes. Consistent with a functional requirement for alx genes in aCNC, alx1; alx3 mutants develop with nearly absent ANC and grossly aberrant hyaloid vasculature and ocular anterior segment, but normal retina. In vivo lineage labeling identified a requirement for alx1 and alx3 during aCNC migration, and transcriptomic analysis suggested oxidative stress response as a key target mechanism of this function. Oxidative stress is a hallmark of fetal alcohol toxicity, and we found increased penetrance of facial and ocular malformations in alx1 mutants exposed to ethanol, consistent with a protective role for alx1 against ethanol toxicity. Collectively, these data demonstrate a conserved role for zebrafish alx genes in controlling ocular and facial development, and a novel role in protecting these key midfacial structures from ethanol toxicity during embryogenesis. These data also reveal novel roles for alx genes in ocular anterior segment formation and vascular development and suggest that retinal deficits in alx mutants may be secondary to aberrant ocular vascularization and anterior segment defects. This study establishes robust zebrafish models for interrogating conserved genetic mechanisms that coordinate facial and ocular development, and for exploring gene--environment interactions relevant to fetal alcohol syndrome.
Collapse
Affiliation(s)
- Baul Yoon
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Pan Yeung
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Nicholas Santistevan
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren E. Bluhm
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Kenta Kawasaki
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Janina Kueper
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
- Institute of Human Genetics, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Richard Dubielzig
- Comparative Ocular Pathology Laboratory of Wisconsin (COPLOW), University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer VanOudenhove
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Justin Cotney
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Yevgenya Grinblat
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
18
|
Lee YY, Cal-Kayitmazbatir S, Francey LJ, Bahiru MS, Hayer KE, Wu G, Zeller MJ, Roberts R, Speers J, Koshalek J, Berres ME, Bittman EL, Hogenesch JB. duper is a null mutation of Cryptochrome 1 in Syrian hamsters. Proc Natl Acad Sci U S A 2022; 119:e2123560119. [PMID: 35471909 PMCID: PMC9170138 DOI: 10.1073/pnas.2123560119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/08/2022] [Indexed: 12/20/2022] Open
Abstract
The duper mutation is a recessive mutation that shortens the period length of the circadian rhythm in Syrian hamsters. These animals show a large phase shift when responding to light pulses. Limited genetic resources for the Syrian hamster (Mesocricetus auratus) presented a major obstacle to cloning duper. This caused the duper mutation to remain unknown for over a decade. In this study, we did a de novo genome assembly of Syrian hamsters with long-read sequencing data from two different platforms, Pacific Biosciences and Oxford Nanopore Technologies. Using two distinct ecotypes and a fast homozygosity mapping strategy, we identified duper as an early nonsense allele of Cryptochrome 1 (Cry1) leading to a short, unstable protein. CRY1 is known as a highly conserved component of the repressive limb of the core circadian clock. The genome assembly and other genomic datasets generated in this study will facilitate the use of the Syrian hamster in biomedical research.
Collapse
Affiliation(s)
- Yin Yeng Lee
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Sibel Cal-Kayitmazbatir
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Lauren J. Francey
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Michael Seifu Bahiru
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003
- Program in Neuroscience & Behavior, University of Massachusetts Amherst, Amherst, MA 01003
| | - Katharina E. Hayer
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104
| | - Gang Wu
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Molly J. Zeller
- University of Wisconsin Biotechnology Center, University of Wisconsin–Madison, Madison, WI 53706
| | - Robyn Roberts
- University of Wisconsin Biotechnology Center, University of Wisconsin–Madison, Madison, WI 53706
| | - James Speers
- University of Wisconsin Biotechnology Center, University of Wisconsin–Madison, Madison, WI 53706
| | - Justin Koshalek
- University of Wisconsin Biotechnology Center, University of Wisconsin–Madison, Madison, WI 53706
| | - Mark E. Berres
- University of Wisconsin Biotechnology Center, University of Wisconsin–Madison, Madison, WI 53706
| | - Eric L. Bittman
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003
- Program in Neuroscience & Behavior, University of Massachusetts Amherst, Amherst, MA 01003
| | - John B. Hogenesch
- Divisions of Human Genetics and Immunobiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| |
Collapse
|
19
|
Echeverry FA, Ijaz S, Pereda AE. Recording Synaptic Transmission from Auditory Mixed Synapses on the Mauthner Cells of Developing Zebrafish. eNeuro 2022; 9:ENEURO.0021-22.2022. [PMID: 35641226 PMCID: PMC9215698 DOI: 10.1523/eneuro.0021-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/14/2022] [Accepted: 05/14/2022] [Indexed: 11/21/2022] Open
Abstract
The Mauthner cells are a pair of large reticulospinal neurons that organize sensory-evoked tail flip responses in fishes. An identifiable group of auditory "mixed" (electrical and chemical) synaptic contacts known as "Large Myelinated Club endings" on these cells have provided a valuable model for the study of synaptic transmission in the vertebrate brain. While most of studies were performed in adult fish, we describe here methods that make possible recording synaptic transmission from these contacts in developing zebrafish, a genetically tractable vertebrate species which is uniquely amenable for combining synaptic physiology with live imaging and behavioral analysis.
Collapse
Affiliation(s)
- Fabio A Echeverry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Sundas Ijaz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
20
|
Scaramella C, Alzagatiti JB, Creighton C, Mankatala S, Licea F, Winter GM, Emtage J, Wisnieski JR, Salazar L, Hussain A, Lee FM, Mammootty A, Mammootty N, Aldujaili A, Runnberg KA, Hernandez D, Zimmerman-Thompson T, Makwana R, Rouvere J, Tahmasebi Z, Zavradyan G, Campbell CS, Komaranchath M, Carmona J, Trevitt J, Glanzman D, Roberts AC. Bisphenol A Exposure Induces Sensory Processing Deficits in Larval Zebrafish during Neurodevelopment. eNeuro 2022; 9:ENEURO.0020-22.2022. [PMID: 35508370 PMCID: PMC9116930 DOI: 10.1523/eneuro.0020-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/10/2022] [Accepted: 04/01/2022] [Indexed: 11/21/2022] Open
Abstract
Because of their ex utero development, relatively simple nervous system, translucency, and availability of tools to investigate neural function, larval zebrafish are an exceptional model for understanding neurodevelopmental disorders and the consequences of environmental toxins. Furthermore, early in development, zebrafish larvae easily absorb chemicals from water, a significant advantage over methods required to expose developing organisms to chemical agents in utero Bisphenol A (BPA) and BPA analogs are ubiquitous environmental toxins with known molecular consequences. All humans have measurable quantities of BPA in their bodies. Most concerning, the level of BPA exposure is correlated with neurodevelopmental difficulties in people. Given the importance of understanding the health-related effects of this common toxin, we have exploited the experimental advantages of the larval zebrafish model system to investigate the behavioral and anatomic effects of BPA exposure. We discovered that BPA exposure early in development leads to deficits in the processing of sensory information, as indicated by BPA's effects on prepulse inhibition (PPI) and short-term habituation (STH) of the C-start reflex. We observed no changes in locomotion, thigmotaxis, and repetitive behaviors (circling). Despite changes in sensory processing, we detected no regional or whole-brain volume changes. Our results show that early BPA exposure can induce sensory processing deficits, as revealed by alterations in simple behaviors that are mediated by a well-defined neural circuit.
Collapse
Affiliation(s)
- Courtney Scaramella
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Joseph B Alzagatiti
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Christopher Creighton
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Samandeep Mankatala
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Fernando Licea
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Gabriel M Winter
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Jasmine Emtage
- Department of Biology, California Institute of Technology, Pasadena, CA 91125
| | - Joseph R Wisnieski
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Luis Salazar
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Anjum Hussain
- Department of Neuroscience, University of California, Riverside, Riverside, CA 92521
| | - Faith M Lee
- Department of Society and Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Asma Mammootty
- Saint Louis University School of Medicine, St. Louis, MO 63104
| | | | - Andrew Aldujaili
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Kristine A Runnberg
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Daniela Hernandez
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | | | - Rikhil Makwana
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Julien Rouvere
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Zahra Tahmasebi
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - Gohar Zavradyan
- Department of Neuroscience, University of California, Riverside, Riverside, CA 92521
| | | | - Meghna Komaranchath
- Department of Biomedical Engineering, Columbia University, New York, NY 10027
| | - Javier Carmona
- Department of Physics, University of California, Los Angeles, Los Angeles, CA 90095
| | - Jennifer Trevitt
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| | - David Glanzman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
- Integrative Center for Learning and Memory, Brain Research Institute, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Adam C Roberts
- Department of Psychology, California State University at Fullerton, Fullerton, CA 92831
| |
Collapse
|
21
|
Abstract
Nervous system assembly relies on a diversity of cellular processes ranging from dramatic tissue reorganization to local, subcellular changes all driven by precise molecular programs. Combined, these processes culminate in an animal's ability to plan and execute behaviors. Animal behavior can, therefore, serve as a functional readout of nervous system development. Benefitting from an expansive and growing set of molecular and imaging tools paired with an ever-growing number of assays of diverse behaviors, the zebrafish system has emerged as an outstanding platform at the intersection of nervous system assembly, plasticity and behavior. Here, we summarize recent advancements in the field, including how developing neural circuits are refined to shape complex behaviors and plasticity.
Collapse
Affiliation(s)
- Jessica C. Nelson
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
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
|
23
|
Marquez-Legorreta E, Constantin L, Piber M, Favre-Bulle IA, Taylor MA, Blevins AS, Giacomotto J, Bassett DS, Vanwalleghem GC, Scott EK. Brain-wide visual habituation networks in wild type and fmr1 zebrafish. Nat Commun 2022; 13:895. [PMID: 35173170 PMCID: PMC8850451 DOI: 10.1038/s41467-022-28299-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Habituation is a form of learning during which animals stop responding to repetitive stimuli, and deficits in habituation are characteristic of several psychiatric disorders. Due to technical challenges, the brain-wide networks mediating habituation are poorly understood. Here we report brain-wide calcium imaging during larval zebrafish habituation to repeated visual looming stimuli. We show that different functional categories of loom-sensitive neurons are located in characteristic locations throughout the brain, and that both the functional properties of their networks and the resulting behavior can be modulated by stimulus saliency and timing. Using graph theory, we identify a visual circuit that habituates minimally, a moderately habituating midbrain population proposed to mediate the sensorimotor transformation, and downstream circuit elements responsible for higher order representations and the delivery of behavior. Zebrafish larvae carrying a mutation in the fmr1 gene have a systematic shift toward sustained premotor activity in this network, and show slower behavioral habituation.
Collapse
Affiliation(s)
- Emmanuel Marquez-Legorreta
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lena Constantin
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Marielle Piber
- School of Medicine, Medical Sciences, and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Itia A Favre-Bulle
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,School of Mathematics and Physics, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Michael A Taylor
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ann S Blevins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean Giacomotto
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD, 4076, Australia.,Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.,Discovery Biology, Griffith University, Brisbane, QLD, 4111, Australia
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Departments of Electrical & Systems Engineering, Physics & Astronomy, Neurology, Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Santa Fe Institute, Santa Fe, NM, 87501, USA
| | - Gilles C Vanwalleghem
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
24
|
Kumari P, Sturgeon M, Bonde G, Cornell RA. Generating Zebrafish RNA-Less Mutant Alleles by Deleting Gene Promoters with CRISPR/Cas9. Methods Mol Biol 2022; 2403:91-106. [PMID: 34913119 PMCID: PMC10136374 DOI: 10.1007/978-1-0716-1847-9_8] [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] [Indexed: 10/19/2022]
Abstract
Danio rerio (zebrafish), traditionally used in forward genetic screens, has in the last decade become a popular model for reverse genetic studies with the introduction of TALENS, zinc finger nucleases, and CRISPR/Cas9. Unexpectedly, homozygous frameshift mutations generated by these tools frequently result in phenotypes that are less penetrant than those seen in embryos injected with antisense morpholino oligonucleotides targeting the same gene. One explanation for the difference is that some frameshift mutations result in nonsense-mediated decay of the gene transcript, a process which can induce expression of homologous genes. This form of genetic compensation, called transcriptional adaptation, does not occur when the mutant allele results in no RNA transcripts being produced from the targeted gene. Such RNA-less mutants can be generated by deleting a gene's promoter using a pair of guide RNAs and Cas9 protein. Here, we present a protocol and use it to generate alleles of arhgap29b and slc41a1 that lack detectable zygotic transcription. In the case of the arhgap29b mutant, an emerging phenotype did not segregate with the promoter deletion mutation, highlighting the potential for off-target mutagenesis with these tools. In summary, this chapter describes a method to generate zebrafish mutants that avoid a form of genetic compensation that occurs in many frameshift mutants.
Collapse
Affiliation(s)
- Priyanka Kumari
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | - Morgan Sturgeon
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Integrated DNA Technologies, Coralville, IA, USA
| | - Gregory Bonde
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | - Robert A Cornell
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
25
|
Burris B, Jensen N, Mokalled MH. Assessment of Swim Endurance and Swim Behavior in Adult Zebrafish. J Vis Exp 2021:10.3791/63240. [PMID: 34842242 PMCID: PMC9226856 DOI: 10.3791/63240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Due to their renowned regenerative capacity, adult zebrafish are a premier vertebrate model to interrogate mechanisms of innate spinal cord regeneration. Following complete transection of their spinal cord, zebrafish extend glial and axonal bridges across severed tissue, regenerate neurons proximal to the lesion, and regain their swim capacities within 8 weeks of injury. Recovery of swim function is thus a central readout for functional spinal cord repair. Here, we describe a set of behavioral assays to quantify zebrafish motor capacity inside an enclosed swim tunnel. The goal of these methods is to provide quantifiable measurements of swim endurance and swim behavior in adult zebrafish. For swim endurance, zebrafish are subjected to a constantly increasing water current velocity until exhaustion, and time at exhaustion is reported. For swim behavior assessment, zebrafish are subjected to low current velocities and swim videos are captured with a dorsal view of the fish. Percent activity, burst frequency, and time spent against the water current provide quantifiable readouts of swim behavior. We quantified swim endurance and swim behavior in wild-type zebrafish before injury and after spinal cord transection. We found that zebrafish lose swim function after spinal cord transection and gradually regain that capacity between 2 and 6 weeks post-injury. The methods described in this study could be applied to neurobehavioral, musculoskeletal, skeletal muscle regeneration, and neural regeneration studies in adult zebrafish.
Collapse
Affiliation(s)
- Brooke Burris
- Department of Developmental Biology, Washington University School of Medicine
| | - Nicholas Jensen
- Department of Developmental Biology, Washington University School of Medicine
| | - Mayssa H Mokalled
- Department of Developmental Biology, Washington University School of Medicine; Center of Regenerative Medicine, Washington University School of Medicine;
| |
Collapse
|
26
|
Brun NR, Panlilio JM, Zhang K, Zhao Y, Ivashkin E, Stegeman JJ, Goldstone JV. Developmental exposure to non-dioxin-like polychlorinated biphenyls promotes sensory deficits and disrupts dopaminergic and GABAergic signaling in zebrafish. Commun Biol 2021; 4:1129. [PMID: 34561524 PMCID: PMC8463681 DOI: 10.1038/s42003-021-02626-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/25/2021] [Indexed: 11/09/2022] Open
Abstract
The most abundant polychlorinated biphenyl (PCB) congeners found in the environment and in humans are neurotoxic. This is of particular concern for early life stages because the exposure of the more vulnerable developing nervous system to neurotoxic chemicals can result in neurobehavioral disorders. In this study, we uncover currently unknown links between PCB target mechanisms and neurobehavioral deficits using zebrafish as a vertebrate model. We investigated the effects of the abundant non-dioxin-like (NDL) congener PCB153 on neuronal morphology and synaptic transmission linked to the proper execution of a sensorimotor response. Zebrafish that were exposed during development to concentrations similar to those found in human cord blood and PCB contaminated sites showed a delay in startle response. Morphological and biochemical data demonstrate that even though PCB153-induced swelling of afferent sensory neurons, the disruption of dopaminergic and GABAergic signaling appears to contribute to PCB-induced motor deficits. A similar delay was observed for other NDL congeners but not for the potent dioxin-like congener PCB126. The effects on important and broadly conserved signaling mechanisms in vertebrates suggest that NDL PCBs may contribute to neurodevelopmental abnormalities in humans and increased selection pressures in vertebrate wildlife.
Collapse
Affiliation(s)
- Nadja R Brun
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, MA, USA.
| | - Jennifer M Panlilio
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Kun Zhang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.,Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China
| | - Yanbin Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.,Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, China
| | - Evgeny Ivashkin
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, USA.,A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - John J Stegeman
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, MA, USA.
| | - Jared V Goldstone
- Department of Biology, Woods Hole Oceanographic Institution, Woods Hole, MA, USA.
| |
Collapse
|
27
|
High Behavioral Variability Mediated by Altered Neuronal Excitability in auts2 Mutant Zebrafish. eNeuro 2021; 8:ENEURO.0493-20.2021. [PMID: 34544758 PMCID: PMC8503961 DOI: 10.1523/eneuro.0493-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 01/28/2023] Open
Abstract
Autism spectrum disorders (ASDs) are characterized by abnormal behavioral traits arising from neural circuit dysfunction. While a number of genes have been implicated in ASDs, in most cases, a clear understanding of how mutations in these genes lead to circuit dysfunction and behavioral abnormality is absent. The autism susceptibility candidate 2 (AUTS2) gene is one such gene, associated with ASDs, intellectual disability and a range of other neurodevelopmental conditions. However, the role of AUTS2 in neural development and circuit function is not at all known. Here, we undertook functional analysis of Auts2a, the main homolog of AUTS2 in zebrafish, in the context of the escape behavior. Escape behavior in wild-type zebrafish is critical for survival and is therefore, reliable, rapid, and has well-defined kinematic properties. auts2a mutant zebrafish are viable, have normal gross morphology and can generate escape behavior with normal kinematics. However, the behavior is unreliable and delayed, with high trial-to-trial variability in the latency. Using calcium imaging we probed the activity of Mauthner neurons during otic vesicle (OV) stimulation and observed lower probability of activation and reduced calcium transients in the mutants. With direct activation of Mauthner by antidromic stimulation, the threshold for activation in mutants was higher than that in wild-type, even when inhibition was blocked. Taken together, these results point to reduced excitability of Mauthner neurons in auts2a mutant larvae leading to unreliable escape responses. Our results show a novel role for Auts2a in regulating neural excitability and reliability of behavior.
Collapse
|
28
|
Corradi L, Filosa A. Neuromodulation and Behavioral Flexibility in Larval Zebrafish: From Neurotransmitters to Circuits. Front Mol Neurosci 2021; 14:718951. [PMID: 34335183 PMCID: PMC8319623 DOI: 10.3389/fnmol.2021.718951] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Animals adapt their behaviors to their ever-changing needs. Internal states, such as hunger, fear, stress, and arousal are important behavioral modulators controlling the way an organism perceives sensory stimuli and reacts to them. The translucent zebrafish larva is an ideal model organism for studying neuronal circuits regulating brain states, owning to the possibility of easy imaging and manipulating activity of genetically identified neurons while the animal performs stereotyped and well-characterized behaviors. The main neuromodulatory circuits present in mammals can also be found in the larval zebrafish brain, with the advantage that they contain small numbers of neurons. Importantly, imaging and behavioral techniques can be combined with methods for generating targeted genetic modifications to reveal the molecular underpinnings mediating the functions of such circuits. In this review we discuss how studying the larval zebrafish brain has contributed to advance our understanding of circuits and molecular mechanisms regulating neuromodulation and behavioral flexibility.
Collapse
Affiliation(s)
- Laura Corradi
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
29
|
Reiss AP, Rankin CH. Gaining an understanding of behavioral genetics through studies of foraging in Drosophila and learning in C. elegans. J Neurogenet 2021; 35:119-131. [PMID: 34151727 DOI: 10.1080/01677063.2021.1928113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The pursuit of understanding behavior has led to investigations of how genes, the environment, and the nervous system all work together to produce and influence behavior, giving rise to a field of research known as behavioral neurogenetics. This review focuses on the research journeys of two pioneers of aspects of behavioral neurogenetic research: Dr. Marla Sokolowski and Dr. Catharine Rankin as examples of how different approaches have been used to understand relationships between genes and behavior. Marla Sokolowski's research is centered around the discovery and analysis of foraging, a gene responsible for the natural behavioral polymorphism of Drosophila melanogaster larvae foraging behavior. Catharine Rankin's work began with demonstrating the ability to learn in Caenorhabditis elegans and then setting out to investigate the mechanisms underlying the "simplest" form of learning, habituation. Using these simple invertebrate organisms both investigators were able to perform in-depth dissections of behavior at genetic and molecular levels. By exploring their research and highlighting their findings we present ways their work has furthered our understanding of behavior and contributed to the field of behavioral neurogenetics.
Collapse
Affiliation(s)
- Aaron P Reiss
- Department of Psychology, University of British Columbia, Vancouver, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Catharine H Rankin
- Department of Psychology, University of British Columbia, Vancouver, Canada.,Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
30
|
Xu J, Casanave R, Guo S. Larval zebrafish display dynamic learning of aversive stimuli in a constant visual surrounding. ACTA ACUST UNITED AC 2021; 28:228-238. [PMID: 34131054 PMCID: PMC8212779 DOI: 10.1101/lm.053425.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022]
Abstract
Balancing exploration and anti-predation are fundamental to the fitness and survival of all animal species from early life stages. How these basic survival instincts drive learning remains poorly understood. Here, using a light/dark preference paradigm with well-controlled luminance history and constant visual surrounding in larval zebrafish, we analyzed intra- and intertrial dynamics for two behavioral components, dark avoidance and center avoidance. We uncover that larval zebrafish display short-term learning of dark avoidance with initial sensitization followed by habituation; they also exhibit long-term learning that is sensitive to trial interval length. We further show that such stereotyped learning patterns is stimulus-specific, as they are not observed for center avoidance. Finally, we demonstrate at individual levels that long-term learning is under homeostatic control. Together, our work has established a novel paradigm to understand learning, uncovered sequential sensitization and habituation, and demonstrated stimulus specificity, individuality, as well as dynamicity in learning.
Collapse
Affiliation(s)
- Jiale Xu
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisico, San Francisco, California 94158, USA
| | - Romelo Casanave
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisico, San Francisco, California 94158, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisico, San Francisco, California 94158, USA.,Program in Human Genetics, University of California at San Francisco, San Francisco, California 94158, USA.,Program in Biological Sciences, University of California at San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
31
|
Meserve JH, Nelson JC, Marsden KC, Hsu J, Echeverry FA, Jain RA, Wolman MA, Pereda AE, Granato M. A forward genetic screen identifies Dolk as a regulator of startle magnitude through the potassium channel subunit Kv1.1. PLoS Genet 2021; 17:e1008943. [PMID: 34061829 PMCID: PMC8195410 DOI: 10.1371/journal.pgen.1008943] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/11/2021] [Accepted: 05/04/2021] [Indexed: 11/19/2022] Open
Abstract
The acoustic startle response is an evolutionarily conserved avoidance behavior. Disruptions in startle behavior, particularly startle magnitude, are a hallmark of several human neurological disorders. While the neural circuitry underlying startle behavior has been studied extensively, the repertoire of genes and genetic pathways that regulate this locomotor behavior has not been explored using an unbiased genetic approach. To identify such genes, we took advantage of the stereotypic startle behavior in zebrafish larvae and performed a forward genetic screen coupled with whole genome analysis. We uncovered mutations in eight genes critical for startle behavior, including two genes encoding proteins associated with human neurological disorders, Dolichol kinase (Dolk), a broadly expressed regulator of the glycoprotein biosynthesis pathway, and the potassium Shaker-like channel subunit Kv1.1. We demonstrate that Kv1.1 and Dolk play critical roles in the spinal cord to regulate movement magnitude during the startle response and spontaneous swim movements. Moreover, we show that Kv1.1 protein is mislocalized in dolk mutants, suggesting they act in a common genetic pathway. Combined, our results identify a diverse set of eight genes, all associated with human disorders, that regulate zebrafish startle behavior and reveal a previously unappreciated role for Dolk and Kv1.1 in regulating movement magnitude via a common genetic pathway.
Collapse
Affiliation(s)
- Joy H. Meserve
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kurt C. Marsden
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jerry Hsu
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Fabio A. Echeverry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Roshan A. Jain
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marc A. Wolman
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alberto E. Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
32
|
Sheets L, Holmgren M, Kindt KS. How Zebrafish Can Drive the Future of Genetic-based Hearing and Balance Research. J Assoc Res Otolaryngol 2021; 22:215-235. [PMID: 33909162 PMCID: PMC8110678 DOI: 10.1007/s10162-021-00798-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Over the last several decades, studies in humans and animal models have successfully identified numerous molecules required for hearing and balance. Many of these studies relied on unbiased forward genetic screens based on behavior or morphology to identify these molecules. Alongside forward genetic screens, reverse genetics has further driven the exploration of candidate molecules. This review provides an overview of the genetic studies that have established zebrafish as a genetic model for hearing and balance research. Further, we discuss how the unique advantages of zebrafish can be leveraged in future genetic studies. We explore strategies to design novel forward genetic screens based on morphological alterations using transgenic lines or behavioral changes following mechanical or acoustic damage. We also outline how recent advances in CRISPR-Cas9 can be applied to perform reverse genetic screens to validate large sequencing datasets. Overall, this review describes how future genetic studies in zebrafish can continue to advance our understanding of inherited and acquired hearing and balance disorders.
Collapse
Affiliation(s)
- Lavinia Sheets
- Department of Otolaryngology-Head & Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Melanie Holmgren
- Department of Otolaryngology-Head & Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Katie S Kindt
- Section On Sensory Cell Development and Function, National Institutes On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
33
|
Onesto MM, Short CA, Rempel SK, Catlett TS, Gomez TM. Growth Factors as Axon Guidance Molecules: Lessons From in vitro Studies. Front Neurosci 2021; 15:678454. [PMID: 34093120 PMCID: PMC8175860 DOI: 10.3389/fnins.2021.678454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Growth cones at the tips of extending axons navigate through developing organisms by probing extracellular cues, which guide them through intermediate steps and onto final synaptic target sites. Widespread focus on a few guidance cue families has historically overshadowed potentially crucial roles of less well-studied growth factors in axon guidance. In fact, recent evidence suggests that a variety of growth factors have the ability to guide axons, affecting the targeting and morphogenesis of growth cones in vitro. This review summarizes in vitro experiments identifying responses and signaling mechanisms underlying axon morphogenesis caused by underappreciated growth factors.
Collapse
Affiliation(s)
| | | | | | | | - Timothy M. Gomez
- Neuroscience Training Program and Cell and Molecular Biology Program, Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
34
|
Lasseigne AM, Echeverry FA, Ijaz S, Michel JC, Martin EA, Marsh AJ, Trujillo E, Marsden KC, Pereda AE, Miller AC. Electrical synaptic transmission requires a postsynaptic scaffolding protein. eLife 2021; 10:e66898. [PMID: 33908867 PMCID: PMC8081524 DOI: 10.7554/elife.66898] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Electrical synaptic transmission relies on neuronal gap junctions containing channels constructed by Connexins. While at chemical synapses neurotransmitter-gated ion channels are critically supported by scaffolding proteins, it is unknown if channels at electrical synapses require similar scaffold support. Here, we investigated the functional relationship between neuronal Connexins and Zonula Occludens 1 (ZO1), an intracellular scaffolding protein localized to electrical synapses. Using model electrical synapses in zebrafish Mauthner cells, we demonstrated that ZO1 is required for robust synaptic Connexin localization, but Connexins are dispensable for ZO1 localization. Disrupting this hierarchical ZO1/Connexin relationship abolishes electrical transmission and disrupts Mauthner cell-initiated escape responses. We found that ZO1 is asymmetrically localized exclusively postsynaptically at neuronal contacts where it functions to assemble intercellular channels. Thus, forming functional neuronal gap junctions requires a postsynaptic scaffolding protein. The critical function of a scaffolding molecule reveals an unanticipated complexity of molecular and functional organization at electrical synapses.
Collapse
Affiliation(s)
| | - Fabio A Echeverry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Sundas Ijaz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | | | - E Anne Martin
- Institute of Neuroscience, University of OregonEugeneUnited States
| | - Audrey J Marsh
- Institute of Neuroscience, University of OregonEugeneUnited States
| | - Elisa Trujillo
- Institute of Neuroscience, University of OregonEugeneUnited States
| | - Kurt C Marsden
- Department of Biological Sciences, NC State UniversityRaleighUnited States
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Adam C Miller
- Institute of Neuroscience, University of OregonEugeneUnited States
| |
Collapse
|
35
|
Alassaf M, Halloran MC. Pregnancy-associated plasma protein-aa regulates endoplasmic reticulum-mitochondria associations. eLife 2021; 10:59687. [PMID: 33759764 PMCID: PMC8024009 DOI: 10.7554/elife.59687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Endoplasmic reticulum (ER) and mitochondria form close physical associations to facilitate calcium transfer, thereby regulating mitochondrial function. Neurons with high metabolic demands, such as sensory hair cells, are especially dependent on precisely regulated ER-mitochondria associations. We previously showed that the secreted metalloprotease pregnancy-associated plasma protein-aa (Pappaa) regulates mitochondrial function in zebrafish lateral line hair cells (Alassaf et al., 2019). Here, we show that pappaa mutant hair cells exhibit excessive and abnormally close ER-mitochondria associations, suggesting increased ER-mitochondria calcium transfer. pappaa mutant hair cells are more vulnerable to pharmacological induction of ER-calcium transfer. Additionally, pappaa mutant hair cells display ER stress and dysfunctional downstream processes of the ER-mitochondria axis including altered mitochondrial morphology and reduced autophagy. We further show that Pappaa influences ER-calcium transfer and autophagy via its ability to stimulate insulin-like growth factor-1 bioavailability. Together our results identify Pappaa as a novel regulator of the ER-mitochondria axis.
Collapse
Affiliation(s)
- Mroj Alassaf
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Department of Neuroscience, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Department of Neuroscience, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| |
Collapse
|
36
|
Bátora D, Zsigmond Á, Lőrincz IZ, Szegvári G, Varga M, Málnási-Csizmadia A. Subcellular Dissection of a Simple Neural Circuit: Functional Domains of the Mauthner-Cell During Habituation. Front Neural Circuits 2021; 15:648487. [PMID: 33828462 PMCID: PMC8019725 DOI: 10.3389/fncir.2021.648487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Abstract
Sensorimotor integration is a pivotal feature of the nervous system for ensuring a coordinated motor response to external stimuli. In essence, such neural circuits can optimize behavioral performance based on the saliency of environmental cues. In zebrafish, habituation of the acoustic startle response (ASR) is a simple behavior integrated into the startle command neurons, called the Mauthner cells. Whereas the essential neuronal components that regulate the startle response have been identified, the principles of how this regulation is integrated at the subcellular regions of the Mauthner cell, which in turn modulate the performance of the behavior, is still not well understood. Here, we reveal mechanistically distinct dynamics of excitatory inputs converging onto the lateral dendrite (LD) and axon initial segment (AIS) of the Mauthner cell by in vivo imaging glutamate release using iGluSnFR, an ultrafast glutamate sensing fluorescent reporter. We find that modulation of glutamate release is dependent on NMDA receptor activity exclusively at the AIS, which is responsible for setting the sensitivity of the startle reflex and inducing a depression of synaptic activity during habituation. In contrast, glutamate-release at the LD is not regulated by NMDA receptors and serves as a baseline component of Mauthner cell activation. Finally, using in vivo calcium imaging at the feed-forward interneuron population component of the startle circuit, we reveal that these cells indeed play pivotal roles in both setting the startle threshold and habituation by modulating the AIS of the Mauthner cell. These results indicate that a command neuron may have several functionally distinct regions to regulate complex aspects of behavior.
Collapse
Affiliation(s)
- Dániel Bátora
- MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary
| | | | | | - Gábor Szegvári
- MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary
| | | | - András Málnási-Csizmadia
- MTA-ELTE Motor Pharmacology Research Group, Budapest, Hungary.,Motorpharma Limited, Budapest, Hungary
| |
Collapse
|
37
|
Klatt Shaw D, Mokalled MH. Efficient CRISPR/Cas9 mutagenesis for neurobehavioral screening in adult zebrafish. G3-GENES GENOMES GENETICS 2021; 11:6179145. [PMID: 33742663 PMCID: PMC8496216 DOI: 10.1093/g3journal/jkab089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/07/2021] [Indexed: 12/22/2022]
Abstract
Adult zebrafish are widely used to interrogate mechanisms of disease development and tissue regeneration. Yet, the prospect of large-scale genetics in adult zebrafish has traditionally faced a host of biological and technical challenges, including inaccessibility of adult tissues to high-throughput phenotyping and the spatial and technical demands of adult husbandry. Here, we describe an experimental pipeline that combines high-efficiency CRISPR/Cas9 mutagenesis with functional phenotypic screening to identify genes required for spinal cord repair in adult zebrafish. Using CRISPR/Cas9 dual-guide ribonucleic proteins, we show selective and combinatorial mutagenesis of 17 genes at 28 target sites with efficiencies exceeding 85% in adult F0 “crispants”. We find that capillary electrophoresis is a reliable method to measure indel frequencies. Using a quantifiable behavioral assay, we identify seven single- or duplicate-gene crispants with reduced functional recovery after spinal cord injury. To rule out off-target effects, we generate germline mutations that recapitulate the crispant regeneration phenotypes. This study provides a platform that combines high-efficiency somatic mutagenesis with a functional phenotypic readout to perform medium- to large-scale genetic studies in adult zebrafish.
Collapse
Affiliation(s)
- Dana Klatt Shaw
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA.,Center of Regenerative Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Mayssa H Mokalled
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA.,Center of Regenerative Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
38
|
Holmgren M, Sheets L. Using the Zebrafish Lateral Line to Understand the Roles of Mitochondria in Sensorineural Hearing Loss. Front Cell Dev Biol 2021; 8:628712. [PMID: 33614633 PMCID: PMC7892962 DOI: 10.3389/fcell.2020.628712] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/23/2020] [Indexed: 01/05/2023] Open
Abstract
Hair cells are the mechanosensory receptors of the inner ear and can be damaged by noise, aging, and ototoxic drugs. This damage often results in permanent sensorineural hearing loss. Hair cells have high energy demands and rely on mitochondria to produce ATP as well as contribute to intracellular calcium homeostasis. In addition to generating ATP, mitochondria produce reactive oxygen species, which can lead to oxidative stress, and regulate cell death pathways. Zebrafish lateral-line hair cells are structurally and functionally analogous to cochlear hair cells but are optically and pharmacologically accessible within an intact specimen, making the zebrafish a good model in which to study hair-cell mitochondrial activity. Moreover, the ease of genetic manipulation of zebrafish embryos allows for the study of mutations implicated in human deafness, as well as the generation of transgenic models to visualize mitochondrial calcium transients and mitochondrial activity in live organisms. Studies of the zebrafish lateral line have shown that variations in mitochondrial activity can predict hair-cell susceptibility to damage by aminoglycosides or noise exposure. In addition, antioxidants have been shown to protect against noise trauma and ototoxic drug–induced hair-cell death. In this review, we discuss the tools and findings of recent investigations into zebrafish hair-cell mitochondria and their involvement in cellular processes, both under homeostatic conditions and in response to noise or ototoxic drugs. The zebrafish lateral line is a valuable model in which to study the roles of mitochondria in hair-cell pathologies and to develop therapeutic strategies to prevent sensorineural hearing loss in humans.
Collapse
Affiliation(s)
- Melanie Holmgren
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Lavinia Sheets
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
39
|
Induction of Short-Term Sensitization by an Aversive Chemical Stimulus in Zebrafish Larvae. eNeuro 2020; 7:ENEURO.0336-19.2020. [PMID: 33004417 PMCID: PMC7729299 DOI: 10.1523/eneuro.0336-19.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Larval zebrafish possess a number of molecular and genetic advantages for rigorous biological analyses of learning and memory. These advantages have motivated the search for novel forms of memory in these animals that can be exploited for understanding the cellular and molecular bases of vertebrate memory formation and consolidation. Here, we report a new form of behavioral sensitization in zebrafish larvae that is elicited by an aversive chemical stimulus [allyl isothiocyanate (AITC)] and that persists for ≥30 min. This form of sensitization is expressed as enhanced locomotion and thigmotaxis, as well as elevated heart rate. To characterize the neural basis of this nonassociative memory, we used transgenic zebrafish expressing the fluorescent calcium indicator GCaMP6 (Chen et al., 2013); because of the transparency of larval zebrafish, we could optically monitor neural activity in the brain of intact transgenic zebrafish before and after the induction of sensitization. We found a distinct brain area, previously linked to locomotion, that exhibited persistently enhanced neural activity following washout of AITC; this enhanced neural activity correlated with the behavioral sensitization. These results establish a novel form of memory in larval zebrafish and begin to unravel the neural basis of this memory.
Collapse
|
40
|
Saberi-Bosari S, Flores KB, San-Miguel A. Deep learning-enabled analysis reveals distinct neuronal phenotypes induced by aging and cold-shock. BMC Biol 2020; 18:130. [PMID: 32967665 PMCID: PMC7510121 DOI: 10.1186/s12915-020-00861-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/01/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Access to quantitative information is crucial to obtain a deeper understanding of biological systems. In addition to being low-throughput, traditional image-based analysis is mostly limited to error-prone qualitative or semi-quantitative assessment of phenotypes, particularly for complex subcellular morphologies. The PVD neuron in Caenorhabditis elegans, which is responsible for harsh touch and thermosensation, undergoes structural degeneration as nematodes age characterized by the appearance of dendritic protrusions. Analysis of these neurodegenerative patterns is labor-intensive and limited to qualitative assessment. RESULTS In this work, we apply deep learning to perform quantitative image-based analysis of complex neurodegeneration patterns exhibited by the PVD neuron in C. elegans. We apply a convolutional neural network algorithm (Mask R-CNN) to identify neurodegenerative subcellular protrusions that appear after cold-shock or as a result of aging. A multiparametric phenotypic profile captures the unique morphological changes induced by each perturbation. We identify that acute cold-shock-induced neurodegeneration is reversible and depends on rearing temperature and, importantly, that aging and cold-shock induce distinct neuronal beading patterns. CONCLUSION The results of this work indicate that implementing deep learning for challenging image segmentation of PVD neurodegeneration enables quantitatively tracking subtle morphological changes in an unbiased manner. This analysis revealed that distinct patterns of morphological alteration are induced by aging and cold-shock, suggesting different mechanisms at play. This approach can be used to identify the molecular components involved in orchestrating neurodegeneration and to characterize the effect of other stressors on PVD degeneration.
Collapse
Affiliation(s)
- Sahand Saberi-Bosari
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, 27695, USA
| | - Kevin B Flores
- Department of Mathematics, North Carolina State University, Raleigh, NC, 27695, USA
| | - Adriana San-Miguel
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
41
|
Rapid Effects of Selection on Brain-wide Activity and Behavior. Curr Biol 2020; 30:3647-3656.e3. [PMID: 32763165 DOI: 10.1016/j.cub.2020.06.086] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/19/2020] [Accepted: 06/24/2020] [Indexed: 11/21/2022]
Abstract
Interindividual variation in behavior and brain activity is universal and provides substrates for natural selection [1-9]. Selective pressures shift the expression of behavioral traits at the population level [10, 11], but the accompanying changes of the underlying neural circuitry have rarely been identified [12, 13]. Selection likely acts through the genetic and/or epigenetic underpinnings of neural activity controlling the selected behavior [14]. Endocrine and neuromodulatory systems participate in behavioral diversity and could provide the substrate for evolutionary modifications [15-21]. Here, we examined brain-wide patterns of activity in larval zebrafish selectively bred over two generations for extreme differences in habituation of the acoustic startle response (ASR) [22]. The ASR is an evolutionarily conserved defensive behavior induced by strong acoustic/vibrational stimuli. ASR habituation shows great individual variability that is stable over days and heritable [4, 22]. Selection for high ASR habituation leads to stronger sound-evoked activation of ASR-processing brain areas. In contrast, animals selected for low habituation displayed stronger spontaneous activity in ASR-processing centers. Ablation of dopaminergic tyrosine hydroxylase (TH) neurons decreased ASR sensitivity. Independently selected ASR habituation lineages link the effect of behavioral selection to dopaminergic caudal hypothalamus (HC) neurons [23]. High ASR habituation co-segregated with decreased spontaneous swimming phenotypes, but visual startle responses were unaffected. Furthermore, high- and low-habituation larvae differed in stress responses as adults. Thus, selective pressure over a couple of generations on ASR habituation behavior is able to induce substantial differences in brain activity, carrying along additional behaviors as piggyback traits that might further affect fitness in the wild. VIDEO ABSTRACT.
Collapse
|
42
|
Nelson JC, Witze E, Ma Z, Ciocco F, Frerotte A, Randlett O, Foskett JK, Granato M. Acute Regulation of Habituation Learning via Posttranslational Palmitoylation. Curr Biol 2020; 30:2729-2738.e4. [PMID: 32502414 PMCID: PMC8446937 DOI: 10.1016/j.cub.2020.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 01/06/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022]
Abstract
Habituation is an adaptive learning process that enables animals to adjust innate behaviors to changes in their environment. Despite its well-documented implications for a wide diversity of behaviors, the molecular and cellular basis of habituation learning is not well understood. Using whole-genome sequencing of zebrafish mutants isolated in an unbiased genetic screen, we identified the palmitoyltransferase Huntingtin interacting protein 14 (Hip14) as a critical regulator of habituation learning. We demonstrate that Hip14 regulates depression of sensory inputs onto an identified hindbrain neuron and provide evidence that Hip14 palmitoylates the Shaker-like K+ voltage-gated channel subunit (Kv1.1), thereby regulating Kv1.1 subcellular localization. Furthermore, we show that, like for Hip14, loss of Kv1.1 leads to habituation deficits and that Hip14 is dispensable in development and instead acts acutely to promote habituation. Combined, these results uncover a previously unappreciated role for acute posttranslational palmitoylation at defined circuit components to regulate learning.
Collapse
Affiliation(s)
- Jessica C Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Eric Witze
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Zhongming Ma
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Francesca Ciocco
- Department of Biology, Haverford College, 370 Lancaster Avenue, Haverford, PA 19041, USA
| | - Abigaile Frerotte
- Department of Biology, Haverford College, 370 Lancaster Avenue, Haverford, PA 19041, USA
| | - Owen Randlett
- Institut NeuroMyoGène, Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon 69008, France
| | - J Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
Liu C, Li S, Noer PR, Kjaer-Sorensen K, Juhl AK, Goldstein A, Ke C, Oxvig C, Duan C. The metalloproteinase Papp-aa controls epithelial cell quiescence-proliferation transition. eLife 2020; 9:e52322. [PMID: 32293560 PMCID: PMC7185994 DOI: 10.7554/elife.52322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/11/2020] [Indexed: 02/06/2023] Open
Abstract
Human patients carrying PAPP-A2 inactivating mutations have low bone mineral density. The underlying mechanisms for this reduced calcification are poorly understood. Using a zebrafish model, we report that Papp-aa regulates bone calcification by promoting Ca2+-transporting epithelial cell (ionocyte) quiescence-proliferation transition. Ionocytes, which are normally quiescent, re-enter the cell cycle under low [Ca2+] stress. Genetic deletion of Papp-aa, but not the closely related Papp-ab, abolished ionocyte proliferation and reduced calcified bone mass. Loss of Papp-aa expression or activity resulted in diminished IGF1 receptor-Akt-Tor signaling in ionocytes. Under low Ca2+ stress, Papp-aa cleaved Igfbp5a. Under normal conditions, however, Papp-aa proteinase activity was suppressed and IGFs were sequestered in the IGF/Igfbp complex. Pharmacological disruption of the IGF/Igfbp complex or adding free IGF1 activated IGF signaling and promoted ionocyte proliferation. These findings suggest that Papp-aa-mediated local Igfbp5a cleavage functions as a [Ca2+]-regulated molecular switch linking IGF signaling to bone calcification by stimulating epithelial cell quiescence-proliferation transition under low Ca2+ stress.
Collapse
Affiliation(s)
- Chengdong Liu
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Shuang Li
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
- College of Ocean and Earth Sciences, Xiamen UniversityXiamenChina
| | | | | | - Anna Karina Juhl
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Allison Goldstein
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Caihuan Ke
- College of Ocean and Earth Sciences, Xiamen UniversityXiamenChina
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Cunming Duan
- Department of Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
44
|
van de Pol ILE, Flik G, Verberk WCEP. Triploidy in zebrafish larvae: Effects on gene expression, cell size and cell number, growth, development and swimming performance. PLoS One 2020; 15:e0229468. [PMID: 32119699 PMCID: PMC7051096 DOI: 10.1371/journal.pone.0229468] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/06/2020] [Indexed: 12/14/2022] Open
Abstract
There is renewed interest in the regulation and consequences of cell size adaptations in studies on understanding the ecophysiology of ectotherms. Here we test if induction of triploidy, which increases cell size in zebrafish (Danio rerio), makes for a good model system to study consequences of cell size. Ideally, diploid and triploid zebrafish should differ in cell size, but should otherwise be comparable in order to be suitable as a model. We induced triploidy by cold shock and compared diploid and triploid zebrafish larvae under standard rearing conditions for differences in genome size, cell size and cell number, development, growth and swimming performance and expression of housekeeping genes and hsp70.1. Triploid zebrafish have larger but fewer cells, and the increase in cell size matched the increase in genome size (+ 50%). Under standard conditions, patterns in gene expression, ontogenetic development and larval growth were near identical between triploids and diploids. However, under demanding conditions (i.e. the maximum swimming velocity during an escape response), triploid larvae performed poorer than their diploid counterparts, especially after repeated stimuli to induce swimming. This result is consistent with the idea that larger cells have less capacity to generate energy, which becomes manifest during repeated physical exertion resulting in increased fatigue. Triploidy induction in zebrafish appears a valid method to increase specifically cell size and this provides a model system to test for consequences of cell size adaptation for the energy budget and swimming performance of this ectothermic vertebrate.
Collapse
Affiliation(s)
- Iris L. E. van de Pol
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, The Netherlands
- * E-mail:
| | - Gert Flik
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, The Netherlands
| | - Wilco C. E. P. Verberk
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
45
|
Fulcher N, Azzopardi E, De Oliveira C, Hudson R, Schormans AL, Zaman T, Allman BL, Laviolette SR, Schmid S. Deciphering midbrain mechanisms underlying prepulse inhibition of startle. Prog Neurobiol 2019; 185:101734. [PMID: 31863802 DOI: 10.1016/j.pneurobio.2019.101734] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/19/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022]
Abstract
Prepulse inhibition (PPI) is an operational measure of sensorimotor gating. Deficits of PPI are a hallmark of schizophrenia and associated with several other psychiatric illnesses such as e.g. autism spectrum disorder, yet the mechanisms underlying PPI are still not fully understood. There is growing evidence contradicting the long-standing hypothesis that PPI is mediated by a short feed-forward midbrain circuitry including inhibitory cholinergic projections from the pedunculopontine tegmental nucleus (PPTg) to the startle pathway. Here, we employed a chemogenetic approach to explore the involvement of the PPTg in general, and cholinergic neurons specifically, in PPI. Activation of inhibitory DREADDs (designer receptors exclusively activated by designer drugs) in the PPTg by systemic administration of clozapine-N-oxide (CNO) disrupted PPI, confirming the involvement of the PPTg in PPI. In contrast, chemogenetic inhibition of specifically cholinergic PPTg neurons had no effect on PPI, but inhibited morphine-induced conditioned place preference (CPP) in the same animals, showing that the DREADDs were effective in modulating behavior. These findings support a functional role of the PPTg and/or neighboring structures in PPI in accordance with previous lesion studies, but also provide strong evidence against the hypothesis that specifically cholinergic PPTg neurons are involved in mediating PPI, implicating rather non-cholinergic midbrain neurons.
Collapse
Affiliation(s)
- Niveen Fulcher
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada
| | - Erin Azzopardi
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Cleusa De Oliveira
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Roger Hudson
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada
| | - Ashley L Schormans
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Tariq Zaman
- University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Brian L Allman
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Steven R Laviolette
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada
| | - Susanne Schmid
- University of Western Ontario, Schulich School of Medicine & Dentistry, Neuroscience Graduate Program, London, ON, N6A 5C1 Canada; University of Western Ontario, Schulich School of Medicine & Dentistry, Department of Anatomy & Cell Biology, London, ON, N6A 5C1 Canada.
| |
Collapse
|
46
|
Bertotto LB, Catron TR, Tal T. Exploring interactions between xenobiotics, microbiota, and neurotoxicity in zebrafish. Neurotoxicology 2019; 76:235-244. [PMID: 31783042 DOI: 10.1016/j.neuro.2019.11.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/18/2022]
Abstract
Susceptibility to xenobiotic exposures is variable. One factor that might account for this is the microbiome, which encompasses all microorganisms, their encoded genes, and associated functions that colonize a host organism. Microbiota harbor the capacity to affect the toxicokinetics and toxicodynamics of xenobiotic exposures. The neurotoxicological effects of environmental chemicals may be modified by intestinal microbes via the microbiota-gut-brain axis. This is a complex, bi-directional signaling pathway between intestinal microbes and the host nervous system. As a model organism, zebrafish are extremely well-placed to illuminate mechanisms by which microbiota modify the developmental neurotoxicity of environmental chemicals. The goal of this review article is to examine the microbiota-gut-brain axis in a toxicological context, specifically focusing on the strengths and weaknesses of the zebrafish model for the investigation of interactions between xenobiotic agents and host-associated microbes. Previous studies describing the relationship between intestinal microbes and host neurodevelopment will be discussed. From a neurotoxicological perspective, studies utilizing zebrafish to assess links between neurotoxicological outcomes and the microbiome are emphasized. Overall, there are major gaps in our understanding the mechanisms by which microbiota interact with xenobiotics to cause or modify host neurotoxicity. In this review, we demonstrate that zebrafish are an ideal model system for studying the complex relationship between chemical exposures, microorganisms, and host neurotoxicological outcomes.
Collapse
Affiliation(s)
- Luísa B Bertotto
- Oak Ridge Institute for Science and Education, US EPA, ORD, NHEERL, ISTD, United States
| | - Tara R Catron
- Oak Ridge Institute for Science and Education, US EPA, ORD, NHEERL, ISTD, United States
| | - Tamara Tal
- US EPA ORD, NHEERL, ISTD, United States.
| |
Collapse
|
47
|
Wang H, Zhu X, Shen J, Zhao EF, He D, Shen H, Liu H, Zhou Y. Quantitative iTRAQ-based proteomic analysis of differentially expressed proteins in aging in human and monkey. BMC Genomics 2019; 20:725. [PMID: 31601169 PMCID: PMC6788010 DOI: 10.1186/s12864-019-6089-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The underlying physiological mechanisms associated with aging are still complex and unclear. As a very important tissue of human body, the circulatory system also plays a very important role in the process of aging. In this study, we use the isobaric tags for relative and absolute quantification (iTRAQ) method to identify differentially expressed proteins in plasma for humans and monkeys between young and aged. Western blotting and behavioral experiment in mice were performed to validate the expression of the candidate protein. RESULTS Between the young / the old humans and the young / the old monkeys 74 and 69 proteins were found to be differently expressed, respectively. For the human samples, these included 38 up-regulated proteins and 36 down-regulated proteins (a fold change ≥1.3 or ≤ 0.667, p value ≤0.05).For the monkey samples, 51 up-regulated proteins and 18 down-regulated proteins (a fold change ≥1.3 or ≤ 0.667, p value ≤0.05). KEGG pathway analysis revealed that phagosome, focal adhesion, ECM-receptor interaction and PI3K/AKT signaling pathway were the most common pathways involved in aging. We found only IGFBP4 protein that existed in up-regulated proteins in aged both for human and monkey. In addition, the differential expression of IGFBP4 was validated by western blot analysis and IGFBP4 treatment mimicked aging-related cognitive dysfunction in mice. CONCLUSIONS This first, the integrated proteomics for the plasma protein of human and monkey reveal one protein-IGFBP4, which was validated by western blotting and behavioral analysis can promote the process of aging. And, iTRAQ analysis showed that proteolytic systems, and inflammatory responses plays an important role in the process of aging. These findings provide a basis for better understanding of the underlying mechanisms involved in aging.
Collapse
Affiliation(s)
- Hao Wang
- Department of Thoracic-Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Xiaoqi Zhu
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Junyan Shen
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - En-Feng Zhao
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
| | - Dajun He
- College of Life Sciences, Key Laboratary of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, Shihezi University, Shihezi, 832003 Xinjiang China
| | - Haitao Shen
- College of Life Sciences, Key Laboratary of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, Shihezi University, Shihezi, 832003 Xinjiang China
| | - Hailiang Liu
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065 China
- College of Life Sciences, Key Laboratary of Xinjiang Phytomedicine Resource and Utilization of Ministry of Education, Shihezi University, Shihezi, 832003 Xinjiang China
| | - Yongxin Zhou
- Department of Thoracic-Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Phosphorylation of Gephyrin in Zebrafish Mauthner Cells Governs Glycine Receptor Clustering and Behavioral Desensitization to Sound. J Neurosci 2019; 39:8988-8997. [PMID: 31558619 DOI: 10.1523/jneurosci.1315-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 01/01/2023] Open
Abstract
The process by which future behavioral responses are shaped by past experiences is one of the central questions in neuroscience. To gain insight into this process at the molecular and cellular levels, we have applied zebrafish larvae to explore behavioral desensitization to sound. A sudden loud noise often evokes a defensive response known as the acoustic startle response (ASR), which is triggered by firing Mauthner cells in teleosts and amphibians. The probability of evoking ASR by suprathreshold sound is reduced after exposure to repetitive auditory stimuli insufficient in amplitude to evoke the ASR (subthreshold). Although it has been suggested that the potentiation of inhibitory glycinergic inputs into Mauthner cell is involved in this desensitization of the ASR, the molecular basis for the potentiation of glycinergic transmission has been unclear. Through the in vivo monitoring of fluorescently-tagged glycine receptors (GlyRs), we here showed that behavioral desensitization to sound in zebrafish is governed by GlyR clustering in Mauthner cells. We further revealed that CaMKII-dependent phosphorylation of the scaffolding protein gephyrin at serine 325 promoted the synaptic accumulation of GlyR on Mauthner neurons through the enhancement of the gephyrin-GlyR binding, which was indispensable for and could induce desensitization of the ASR. Our study demonstrates an essential molecular and cellular basis of sound-induced receptor dynamics and thus of behavioral desensitization to sound.SIGNIFICANCE STATEMENT Behavioral desensitization in the acoustic startle response of fish is known to involve the potentiation of inhibitory glycinergic input to the Mauthner cell, which is a command neuron for the acoustic startle response. However, the molecular and cellular basis for this potentiation has been unknown. Here we show that an increase in glycine receptor (GlyR) clustering at synaptic sites on zebrafish Mauthner cells is indispensable for and could induce desensitization. Furthermore, we demonstrate that CaMKII-mediated phosphorylation of the scaffolding protein gephyrin promotes GlyR clustering by increasing the binding between the β-loop of GlyRs and gephyrin. Thus, the phosphorylation of gephyrin is a key event which accounts for the potentiation of inhibitory glycinergic inputs observed during sound-evoked behavioral desensitization.
Collapse
|
49
|
Marsden KC, Jain RA, Wolman MA, Echeverry FA, Nelson JC, Hayer KE, Miltenberg B, Pereda AE, Granato M. A Cyfip2-Dependent Excitatory Interneuron Pathway Establishes the Innate Startle Threshold. Cell Rep 2019; 23:878-887. [PMID: 29669291 PMCID: PMC6642828 DOI: 10.1016/j.celrep.2018.03.095] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/13/2017] [Accepted: 03/20/2018] [Indexed: 11/18/2022] Open
Abstract
Sensory experiences dynamically modify whether animals respond to a given stimulus, but it is unclear how innate behavioral thresholds are established. Here, we identify molecular and circuit-level mechanisms underlying the innate threshold of the zebrafish startle response. From a forward genetic screen, we isolated five mutant lines with reduced innate startle thresholds. Using whole-genome sequencing, we identify the causative mutation for one line to be in the fragile X mental retardation protein (FMRP)-interacting protein cyfip2. We show that cyfip2 acts independently of FMRP and that reactivation of cyfip2 restores the baseline threshold after phenotype onset. Finally, we show that cyfip2 regulates the innate startle threshold by reducing neural activity in a small group of excitatory hindbrain interneurons. Thus, we identify a selective set of genes critical to establishing an innate behavioral threshold and uncover a circuit-level role for cyfip2 in this process. Using forward genetics, electrophysiology, and combined behavior and Ca2+ imaging in zebrafish, Marsden et al. show that cyfip2 regulates the acoustic startle threshold by controlling the activity of excitatory spiral fiber interneurons.
Collapse
Affiliation(s)
- Kurt C Marsden
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 1157 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104, USA; Department of Biological Sciences, W.M. Keck Center for Behavioral Biology, North Carolina State University, 127 David Clark Labs, 100 Brooks Ave., Raleigh, NC 27607, USA
| | - Roshan A Jain
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 1157 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104, USA; Department of Biology, Haverford College, S108 Sharpless Hall, 370 Lancaster Ave., Haverford, PA 19041, USA
| | - Marc A Wolman
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 1157 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104, USA; Department of Zoology, 213 Zoology Research Building, 1117 West Johnson St., University of Wisconsin, Madison, WI 53706, USA
| | - Fabio A Echeverry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 431 Rose F. Kennedy Center, 1410 Pelham Parkway South, Bronx, NY 10461, USA
| | - Jessica C Nelson
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 1157 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104, USA
| | - Katharina E Hayer
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ben Miltenberg
- Department of Biology, Haverford College, S108 Sharpless Hall, 370 Lancaster Ave., Haverford, PA 19041, USA
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 431 Rose F. Kennedy Center, 1410 Pelham Parkway South, Bronx, NY 10461, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 1157 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Alassaf M, Daykin EC, Mathiaparanam J, Wolman MA. Pregnancy-associated plasma protein-aa supports hair cell survival by regulating mitochondrial function. eLife 2019; 8:47061. [PMID: 31205004 PMCID: PMC6594750 DOI: 10.7554/elife.47061] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022] Open
Abstract
To support cell survival, mitochondria must balance energy production with oxidative stress. Inner ear hair cells are particularly vulnerable to oxidative stress; thus require tight mitochondrial regulation. We identified a novel molecular regulator of the hair cells’ mitochondria and survival: Pregnancy-associated plasma protein-aa (Pappaa). Hair cells in zebrafish pappaa mutants exhibit mitochondrial defects, including elevated mitochondrial calcium, transmembrane potential, and reactive oxygen species (ROS) production and reduced antioxidant expression. In pappaa mutants, hair cell death is enhanced by stimulation of mitochondrial calcium or ROS production and suppressed by a mitochondrial ROS scavenger. As a secreted metalloprotease, Pappaa stimulates extracellular insulin-like growth factor 1 (IGF1) bioavailability. We found that the pappaa mutants’ enhanced hair cell loss can be suppressed by stimulation of IGF1 availability and that Pappaa-IGF1 signaling acts post-developmentally to support hair cell survival. These results reveal Pappaa as an extracellular regulator of hair cell survival and essential mitochondrial function.
Collapse
Affiliation(s)
- Mroj Alassaf
- Department of Integrative Biology, University of Wisconsin, Madison, United States.,Neuroscience Training Program, University of Wisconsin, Madison, United States
| | - Emily C Daykin
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| | - Jaffna Mathiaparanam
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| | - Marc A Wolman
- Department of Integrative Biology, University of Wisconsin, Madison, United States
| |
Collapse
|