1
|
Wasser-Bennett G, Brown AR, Maynard SK, Owen SF, Tyler CR. Critical insights into the potential risks of antipsychotic drugs to fish, including through effects on behaviour. Biol Rev Camb Philos Soc 2025. [PMID: 40355132 DOI: 10.1111/brv.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
Antipsychotic drugs (APDs) are a diverse class of neuroactive pharmaceuticals increasingly detected in surface and ground waters globally. Some APDs are classified as posing a high environmental risk, due, in part, to their tendency to bioaccumulate in wildlife, including fish. Additional risk drivers for APDs relate to their behavioural effects, potentially impacting fitness outcomes. However, standard ecotoxicological tests used in environmental risk assessment (ERA) do not currently account for these mechanisms. In this review, we critically appraise the environmental risks of APDs to fish. We begin by reading-across from human and mammalian effects data to standard ecotoxicological effects endpoints in fish. We then explore the wide range of behaviours suitable for ecotoxicological assessment of APDs (and other neuroactive) pharmaceuticals, principally through laboratory studies with zebrafish, and assess the potential for using these behavioural phenotypes to predict adverse individual- and population-level outcomes in wild fish, taking into account phenotypic plasticity. Next, we illustrate the advantages and challenges of measuring and applying behavioural endpoints for fish, including within current regulatory risk assessments. In our final analysis, the implications of relying on apical endpoints for ERA of neuroactive drugs (including APDs) are assessed and recommendations provided for the development of a more refined and tailored mechanistic approach, which would enable more robust assessment of their environmental risk(s).
Collapse
Affiliation(s)
- Gabrielle Wasser-Bennett
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, Devon, UK
| | - A Ross Brown
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, Devon, UK
| | - Samuel K Maynard
- AstraZeneca, Global Environment, Macclesfield, Cheshire, SK10 2NA, UK
| | - Stewart F Owen
- AstraZeneca, Global Environment, Macclesfield, Cheshire, SK10 2NA, UK
| | - Charles R Tyler
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, Devon, UK
| |
Collapse
|
2
|
Heagy FK, Clements KN, Adams CL, Blain E, Issa FA. Socially induced plasticity of the posterior tuberculum and motor behavior in zebrafish (Danio rerio). J Exp Biol 2024; 227:jeb248148. [PMID: 39422204 PMCID: PMC11626077 DOI: 10.1242/jeb.248148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Social dominance is prevalent throughout the animal kingdom. It facilitates the stabilization of social relationships and allows animals to divide resources according to social rank. Zebrafish form stable dominance relationships that consist of dominants and subordinates. Although social status-dependent differences in behavior must arise as a result of neural plasticity, mechanisms by which neural circuits are reconfigured to cope with social dominance are poorly described. Here, we describe how the posterior tuberculum nucleus (PTN), which integrates sensory social information to modulate spinal motor circuits, is morphologically and functionally influenced by social status. We combined non-invasive behavioral monitoring of motor activity (startle escape and swim) and histological approaches to investigate how social dominance affects the morphological structure, axosomatic synaptic connectivity and functional activity of the PTN in relation to changes in motor behavior. We show that dopaminergic cell number significantly increases in dominants compared with subordinates, while PTN synaptic interconnectivity, demonstrated with PSD-95 expression, is higher in subordinates than in dominants. Secondly, these socially induced morphological differences emerge after 1 week of dominance formation and correlate with differences in cellular activities illustrated with higher phosphor-S6 ribosomal protein expression in dominants compared with subordinates. Thirdly, these morphological differences are reversible as the social environment evolves and correlate with adaptations in startle escape and swim behaviors. Our results provide new insights into the neural bases of social behavior that may be applicable to other social species with similar structural and functional organization.
Collapse
Affiliation(s)
- Faith K. Heagy
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Katie N. Clements
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Carrie L. Adams
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Elena Blain
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Fadi A. Issa
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| |
Collapse
|
3
|
Ryczko D, Dubuc R. Dopamine control of downstream motor centers. Curr Opin Neurobiol 2023; 83:102785. [PMID: 37774481 DOI: 10.1016/j.conb.2023.102785] [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: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/26/2023] [Indexed: 10/01/2023]
Abstract
The role of dopamine in the control of movement is traditionally associated with ascending projections to the basal ganglia. However, more recently descending dopaminergic pathways projecting to downstream brainstem motor circuits were discovered. In lampreys, salamanders, and rodents, these include projections to the downstream Mesencephalic Locomotor Region (MLR), a brainstem region controlling locomotion. Such descending dopaminergic projections could prime brainstem networks controlling movement. Other descending dopaminergic projections have been shown to reach reticulospinal cells involved in the control of locomotion. In addition, dopamine directly modulates the activity of interneurons and motoneurons. Beyond locomotion, dopaminergic inputs modulate visuomotor transformations within the optic tectum (mammalian superior colliculus). Loss of descending dopaminergic inputs will likely contribute to pathological conditions such as in Parkinson's disease.
Collapse
Affiliation(s)
- Dimitri Ryczko
- Département de Pharmacologie-Physiologie, Université de Sherbrooke, Sherbrooke, Québec, Canada; Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada; Neurosciences Sherbrooke, Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Sherbrooke, Canada.
| | - Réjean Dubuc
- Groupe de Recherche en Activité Physique Adaptée, Département des Sciences de l'Activité Physique, Université du Québec à Montréal, Montréal, Québec, Canada; Groupe de recherche sur la Signalisation Neurale et la Circuiterie, Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
4
|
Chen F, Köhler M, Cucun G, Takamiya M, Kizil C, Cosacak MI, Rastegar S. sox1a:eGFP transgenic line and single-cell transcriptomics reveal the origin of zebrafish intraspinal serotonergic neurons. iScience 2023; 26:107342. [PMID: 37529101 PMCID: PMC10387610 DOI: 10.1016/j.isci.2023.107342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/03/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Abstract
Sox transcription factors are crucial for vertebrate nervous system development. In zebrafish embryo, sox1 genes are expressed in neural progenitor cells and neurons of ventral spinal cord. Our recent study revealed that the loss of sox1a and sox1b function results in a significant decline of V2 subtype neurons (V2s). Using single-cell RNA sequencing, we analyzed the transcriptome of sox1a lineage progenitors and neurons in the zebrafish spinal cord at four time points during embryonic development, employing the Tg(sox1a:eGFP) line. In addition to previously characterized sox1a-expressing neurons, we discovered the expression of sox1a in late-developing intraspinal serotonergic neurons (ISNs). Developmental trajectory analysis suggests that ISNs arise from lateral floor plate (LFP) progenitor cells. Pharmacological inhibition of the Notch signaling pathway revealed its role in negatively regulating LFP progenitor cell differentiation into ISNs. Our findings highlight the zebrafish LFP as a progenitor domain for ISNs, alongside known Kolmer-Agduhr (KA) and V3 interneurons.
Collapse
Affiliation(s)
- Fushun Chen
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Melina Köhler
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Gokhan Cucun
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Masanari Takamiya
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Caghan Kizil
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Tatzberg 41, 01307 Dresden, Germany
- Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY 10032, USA
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Tatzberg 41, 01307 Dresden, Germany
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Campus North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
5
|
Morash MG, Nixon J, Shimoda LMN, Turner H, Stokes AJ, Small-Howard AL, Ellis LD. Identification of minimum essential therapeutic mixtures from cannabis plant extracts by screening in cell and animal models of Parkinson's disease. Front Pharmacol 2022; 13:907579. [PMID: 36278152 PMCID: PMC9586206 DOI: 10.3389/fphar.2022.907579] [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: 03/29/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Medicinal cannabis has shown promise for the symptomatic treatment of Parkinson's disease (PD), but patient exposure to whole plant mixtures may be undesirable due to concerns around safety, consistency, regulatory issues, and psychoactivity. Identification of a subset of components responsible for the potential therapeutic effects within cannabis represents a direct path forward for the generation of anti-PD drugs. Using an in silico database, literature reviews, and cell based assays, GB Sciences previously identified and patented a subset of five cannabinoids and five terpenes that could potentially recapitulate the anti-PD attributes of cannabis. While this work represents a critical step towards harnessing the anti-PD capabilities of cannabis, polypharmaceutical drugs of this complexity may not be feasible as therapeutics. In this paper, we utilize a reductionist approach to identify minimal essential mixtures (MEMs) of these components that are amenable to pharmacological formulation. In the first phase, cell-based models revealed that the cannabinoids had the most significant positive effects on neuroprotection and dopamine secretion. We then evaluated the ability of combinations of these cannabinoids to ameliorate a 6-hydroxydopmamine (OHDA)-induced change in locomotion in larval zebrafish, which has become a well-established PD disease model. Equimolar mixtures that each contained three cannabinoids were able to significantly reverse the OHDA mediated changes in locomotion and other advanced metrics of behavior. Additional screening of sixty-three variations of the original cannabinoid mixtures identified five highly efficacious mixtures that outperformed the original equimolar cannabinoid MEMs and represent the most attractive candidates for therapeutic development. This work highlights the strength of the reductionist approach for the development of ratio-controlled, cannabis mixture-based therapeutics for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
| | - Jessica Nixon
- National Research Council of Canada, Halifax, NS, Canada
| | - Lori M. N. Shimoda
- Laboratory of Immunology and Signal Transduction, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI, United States
| | - Helen Turner
- Laboratory of Immunology and Signal Transduction, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI, United States
| | - Alexander J. Stokes
- Laboratory of Experimental Medicine, John A Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | | | - Lee D. Ellis
- National Research Council of Canada, Halifax, NS, Canada
| |
Collapse
|
6
|
Hong X, Chen R, Zhang L, Yan L, Xin J, Li J, Zha J. Long-Term Exposure to SSRI Citalopram Induces Neurotoxic Effects in Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:12380-12390. [PMID: 35985052 DOI: 10.1021/acs.est.2c01514] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Residual antidepressants are of increasing concern worldwide, yet critical information on their long-term neurotoxic impacts on nontarget aquatic animals is lacking. Here, we investigated the long-term effects (from 0 to 150 days postfertilization) of the selective serotonin reuptake inhibitor citalopram (0.1-100 μg/L) on motor function, learning, and memory in zebrafish over two generations and explored the reversibility of the effect in F1 larvae. Unlike F0+ larvae, we found that F1+ larvae displayed decreased sensorimotor performance when continuously exposed to citalopram at 100 μg/L. No adverse effects were found in F1- larvae after they were transferred to a clean medium. Whole-mount immunofluorescence assays suggested that the motor impairments were related to axonal projections of the spinal motor neurons (MNs). For F0+ adults, long-term citalopram exposure mainly caused male-specific declines in motor, learning, and memory performance. Analysis of serotonergic and cholinergic MNs revealed no significant changes in the male zebrafish spinal cord. In contrast, the number of glutamatergic spinal MNs decreased, likely associated with the impairment of motor function. Additionally, treatment with 100 μg/L citalopram significantly reduced the number of dopaminergic neurons, but no significant neuronal apoptosis was observed in the adult telencephalon. Overall, this study provides neurobehavioral evidence and novel insights into the neurotoxic mechanisms of long-term citalopram exposure and may facilitate the assessment of the environmental and health risks posed by citalopram-containing antidepressant drugs.
Collapse
Affiliation(s)
- Xiangsheng Hong
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Rui Chen
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Le Zhang
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Yan
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiajing Xin
- Department of Public Health, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, China
| | - Jiasu Li
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jinmiao Zha
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
7
|
Huang W, Wu T, Wu K. Zebrafish (Danio rerio): A potential model to assess developmental toxicity of ketamine. CHEMOSPHERE 2022; 291:133033. [PMID: 34822872 DOI: 10.1016/j.chemosphere.2021.133033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023]
Abstract
Ketamine is a non-competitive antagonist of NMDA glutamate receptor. It is used as an anesthetic, analgesic, sedative, and anti-depressive agent in clinical practice and also an illegal recreational drug. The increasing use has contributed to the measurable levels of ketamine in both wastewaters and hospital effluents, thereby classified as an emergent contaminant. Lately, the potential toxicity of ketamine has raised serious concerns about its iatrogenic or illicit use during pregnancy, neonatal and childhood stages. However, to assess its long-term toxicity potentially by the use of early life stages in human and rodents is limited. In this regard, the zebrafish has been considered as excellent model organism for biosafety assessments of ketamine due to it boasts an in vivo model with the advantages of an in vitro assay. In this review, we summarize the current understanding of the reported toxicity studies with ketamine in early life stage of zebrafish. The adverse effects of ketamine are known to cause overall developmental and multi-organ toxicity, including cardio-, neuro-, and skeletal toxicity. Furthermore, multiple mechanisms are found to be responsible for perpetrating toxicity of ketamine. The current findings confluence to emphasize the zebrafish embryo as an appealing model system for developmental toxicity testing in higher vertebrates.
Collapse
Affiliation(s)
- Wenlong Huang
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, PR China
| | - Tianjie Wu
- Department of Anaesthesiology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou, 515041, Guangdong, PR China
| | - Kusheng Wu
- Department of Preventive Medicine, Shantou University Medical College, Shantou, 515041, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Breast Cancer Diagnosis and Treatment, Shantou, 515041, Guangdong, PR China
| |
Collapse
|
8
|
Altenhofen S, Bonan CD. Zebrafish as a tool in the study of sleep and memory-related disorders. Curr Neuropharmacol 2021; 20:540-549. [PMID: 34254919 DOI: 10.2174/1570159x19666210712141041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/23/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
Sleep is an evolutionarily conserved phenomenon, being an essential biological necessity for the learning process and memory consolidation. The brain displays two types of electrical activity during sleep: slow-wave activity or non-rapid eye movement (NREM) sleep and desynchronized brain wave activity or rapid eye movement (REM) sleep. There are many theories about "Why we need to sleep?" among them the synaptic homeostasis. This theory proposes that the role of sleep is the restoration of synaptic homeostasis, which is destabilized by synaptic strengthening triggered by learning during waking and by synaptogenesis during development. Sleep diminishes the plasticity load on neurons and other cells to normalize synaptic strength. In contrast, it re-establishes neuronal selectivity and the ability to learn, leading to the consolidation and integration of memories. The use of zebrafish as a tool to assess sleep and its disorders is growing, although sleep in this animal is not yet divided, for example, into REM and NREM states. However, zebrafish are known to have a regulated daytime circadian rhythm. Their sleep state is characterized by periods of inactivity accompanied by an increase in arousal threshold, preference for resting place, and the "rebound sleep effect" phenomenon, which causes an increased slow-wave activity after a forced waking period. In addition, drugs known to modulate sleep, such as melatonin, nootropics, and nicotine, have been tested in zebrafish. In this review, we discuss the use of zebrafish as a model to investigate sleep mechanisms and their regulation, demonstrating this species as a promising model for sleep research.
Collapse
Affiliation(s)
- Stefani Altenhofen
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celulare Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celulare Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, porto Alegre, RS, Brazil
| |
Collapse
|
9
|
Orr SA, Ahn S, Park C, Miller TH, Kassai M, Issa FA. Social Experience Regulates Endocannabinoids Modulation of Zebrafish Motor Behaviors. Front Behav Neurosci 2021; 15:668589. [PMID: 34045945 PMCID: PMC8144649 DOI: 10.3389/fnbeh.2021.668589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
Social status-dependent modulation of neural circuits has been investigated extensively in vertebrate and invertebrate systems. However, the effects of social status on neuromodulatory systems that drive motor activity are poorly understood. Zebrafish form a stable social relationship that consists of socially dominant and subordinate animals. The locomotor behavior patterns differ according to their social ranks. The sensitivity of the Mauthner startle escape response in subordinates increases compared to dominants while dominants increase their swimming frequency compared to subordinates. Here, we investigated the role of the endocannabinoid system (ECS) in mediating these differences in motor activities. We show that brain gene expression of key ECS protein pathways are socially regulated. Diacylglycerol lipase (DAGL) expression significantly increased in dominants and significantly decreased in subordinates relative to controls. Moreover, brain gene expression of the cannabinoid 1 receptor (CB1R) was significantly increased in subordinates relative to controls. Secondly, increasing ECS activity with JZL184 reversed swimming activity patterns in dominant and subordinate animals. JZL184 did not affect the sensitivity of the startle escape response in dominants while it was significantly reduced in subordinates. Thirdly, blockage of CB1R function with AM-251 had no effect on dominants startle escape response sensitivity, but startle sensitivity was significantly reduced in subordinates. Additionally, AM-251 did not affect swimming activities in either social phenotypes. Fourthly, we demonstrate that the effects of ECS modulation of the startle escape circuit is mediated via the dopaminergic system specifically via the dopamine D1 receptor. Finally, our empirical results complemented with neurocomputational modeling suggest that social status influences the ECS to regulate the balance in synaptic strength between excitatory and inhibitory inputs to control the excitability of motor behaviors. Collectively, this study provides new insights of how social factors impact nervous system function to reconfigure the synergistic interactions of neuromodulatory pathways to optimize motor output.
Collapse
Affiliation(s)
- Stephen A Orr
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Sungwoo Ahn
- Department of Mathematics, East Carolina University, Greenville, NC, United States
| | - Choongseok Park
- Department of Mathematics, North Carolina A&T State University, Greensboro, NC, United States
| | - Thomas H Miller
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Miki Kassai
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Fadi A Issa
- Department of Biology, East Carolina University, Greenville, NC, United States
| |
Collapse
|
10
|
James DM, Davidson EA, Yanes J, Moshiree B, Dallman JE. The Gut-Brain-Microbiome Axis and Its Link to Autism: Emerging Insights and the Potential of Zebrafish Models. Front Cell Dev Biol 2021; 9:662916. [PMID: 33937265 PMCID: PMC8081961 DOI: 10.3389/fcell.2021.662916] [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: 02/01/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
Research involving autism spectrum disorder (ASD) most frequently focuses on its key diagnostic criteria: restricted interests and repetitive behaviors, altered sensory perception, and communication impairments. These core criteria, however, are often accompanied by numerous comorbidities, many of which result in severe negative impacts on quality of life, including seizures, epilepsy, sleep disturbance, hypotonia, and GI distress. While ASD is a clinically heterogeneous disorder, gastrointestinal (GI) distress is among the most prevalent co-occurring symptom complex, manifesting in upward of 70% of all individuals with ASD. Consistent with this high prevalence, over a dozen family foundations that represent genetically distinct, molecularly defined forms of ASD have identified GI symptoms as an understudied area with significant negative impacts on quality of life for both individuals and their caregivers. Moreover, GI symptoms are also correlated with more pronounced irritability, social withdrawal, stereotypy, hyperactivity, and sleep disturbances, suggesting that they may exacerbate the defining behavioral symptoms of ASD. Despite these facts (and to the detriment of the community), GI distress remains largely unaddressed by ASD research and is frequently regarded as a symptomatic outcome rather than a potential contributory factor to the behavioral symptoms. Allowing for examination of both ASD's impact on the central nervous system (CNS) as well as its impact on the GI tract and the associated microbiome, the zebrafish has recently emerged as a powerful tool to study ASD. This is in no small part due to the advantages zebrafish present as a model system: their precocious development, their small transparent larval form, and their parallels with humans in genetics and physiology. While ASD research centered on the CNS has leveraged these advantages, there has been a critical lack of GI-centric ASD research in zebrafish models, making a holistic view of the gut-brain-microbiome axis incomplete. Similarly, high-throughput ASD drug screens have recently been developed but primarily focus on CNS and behavioral impacts while potential GI impacts have not been investigated. In this review, we aim to explore the great promise of the zebrafish model for elucidating the roles of the gut-brain-microbiome axis in ASD.
Collapse
Affiliation(s)
- David M. James
- Department of Biology, University of Miami, Coral Gables, FL, United States
| | | | - Julio Yanes
- Department of Biology, University of Miami, Coral Gables, FL, United States
| | - Baharak Moshiree
- Department of Gastroenterology and Hepatology, Atrium Health, Charlotte, NC, United States
| | - Julia E. Dallman
- Department of Biology, University of Miami, Coral Gables, FL, United States
| |
Collapse
|
11
|
Ryczko D, Grätsch S, Alpert MH, Cone JJ, Kasemir J, Ruthe A, Beauséjour PA, Auclair F, Roitman MF, Alford S, Dubuc R. Descending Dopaminergic Inputs to Reticulospinal Neurons Promote Locomotor Movements. J Neurosci 2020; 40:8478-8490. [PMID: 32998974 PMCID: PMC7605428 DOI: 10.1523/jneurosci.2426-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 01/25/2023] Open
Abstract
Meso-diencephalic dopaminergic neurons are known to modulate locomotor behaviors through their ascending projections to the basal ganglia, which in turn project to the mesencephalic locomotor region, known to control locomotion in vertebrates. In addition to their ascending projections, dopaminergic neurons were found to increase locomotor movements through direct descending projections to the mesencephalic locomotor region and spinal cord. Intriguingly, fibers expressing tyrosine hydroxylase (TH), the rate-limiting enzyme of dopamine synthesis, were also observed around reticulospinal neurons of lampreys. We now examined the origin and the role of this innervation. Using immunofluorescence and tracing experiments, we found that fibers positive for dopamine innervate reticulospinal neurons in the four reticular nuclei of lampreys. We identified the dopaminergic source using tracer injections in reticular nuclei, which retrogradely labeled dopaminergic neurons in a caudal diencephalic nucleus (posterior tuberculum [PT]). Using voltammetry in brain preparations isolated in vitro, we found that PT stimulation evoked dopamine release in all four reticular nuclei, but not in the spinal cord. In semi-intact preparations where the brain is accessible and the body moves, PT stimulation evoked swimming, and injection of a D1 receptor antagonist within the middle rhombencephalic reticular nucleus was sufficient to decrease reticulospinal activity and PT-evoked swimming. Our study reveals that dopaminergic neurons have access to command neurons that integrate sensory and descending inputs to activate spinal locomotor neurons. As such, our findings strengthen the idea that dopamine can modulate locomotor behavior both via ascending projections to the basal ganglia and through descending projections to brainstem motor circuits.SIGNIFICANCE STATEMENT Meso-diencephalic dopaminergic neurons play a key role in modulating locomotion by releasing dopamine in the basal ganglia, spinal networks, and the mesencephalic locomotor region, a brainstem region that controls locomotion in a graded fashion. Here, we report in lampreys that dopaminergic neurons release dopamine in the four reticular nuclei where reticulospinal neurons are located. Reticulospinal neurons integrate sensory and descending suprareticular inputs to control spinal interneurons and motoneurons. By directly modulating the activity of reticulospinal neurons, meso-diencephalic dopaminergic neurons control the very last instructions sent by the brain to spinal locomotor circuits. Our study reports on a new direct descending dopaminergic projection to reticulospinal neurons that modulates locomotor behavior.
Collapse
Affiliation(s)
- Dimitri Ryczko
- Department of Neuroscience, Université de Montréal, Montréal, Québec H3C 3J7, Canada
- Department of Pharmacology-Physiology, Université de Sherbrooke, Sherbrooke J1H 5N4, Québec Canada
- Centre de recherche du CHUS, Sherbrooke, J1H 5N4, Québec, Canada
| | - Swantje Grätsch
- Department of Neuroscience, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Michael H Alpert
- Department of Biological Sciences, University of Illinois at Chicago, Chicago IL 60607, Illinois
| | - Jackson J Cone
- Department of Psychology, University of Illinois at Chicago, Chicago IL 60607, Illinois
| | - Jacquelin Kasemir
- Department of Neuroscience, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Angelina Ruthe
- Department of Neuroscience, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | | | - François Auclair
- Department of Neuroscience, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Mitchell F Roitman
- Department of Psychology, University of Illinois at Chicago, Chicago IL 60607, Illinois
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago IL 60612-7308, Illinois
| | - Réjean Dubuc
- Department of Neuroscience, Université de Montréal, Montréal, Québec H3C 3J7, Canada
- Groupe de Recherche en Activité Physique Adaptée, Department of Exercise Science, Université du Québec à Montréal, Montréal, Québec H3C 3P8, Canada
| |
Collapse
|
12
|
Barrios JP, Wang WC, England R, Reifenberg E, Douglass AD. Hypothalamic Dopamine Neurons Control Sensorimotor Behavior by Modulating Brainstem Premotor Nuclei in Zebrafish. Curr Biol 2020; 30:4606-4618.e4. [PMID: 33007241 DOI: 10.1016/j.cub.2020.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/26/2020] [Accepted: 09/02/2020] [Indexed: 01/10/2023]
Abstract
Dopamine (DA)-producing neurons are critically involved in the production of motor behaviors in multiple circuits that are conserved from basal vertebrates to mammals. Although there is increasing evidence that DA neurons in the hypothalamus play a locomotor role, their precise contributions to behavior and the circuit mechanisms by which they are achieved remain unclear. Here, we demonstrate that tyrosine-hydroxylase-2-expressing (th2+) DA neurons in the zebrafish hypothalamus fire phasic bursts of activity to acutely promote swimming and modulate audiomotor behaviors on fast timescales. Their anatomy and physiology reveal two distinct functional DA modules within the hypothalamus. The first comprises an interconnected set of cerebrospinal-fluid-contacting DA nuclei surrounding the 3rd ventricle, which lack distal projections outside of the hypothalamus and influence locomotion through unknown means. The second includes neurons in the preoptic nucleus, which send long-range projections to targets throughout the brain, including the mid- and hindbrain, where they activate premotor circuits involved in swimming and sensorimotor integration. These data suggest a broad regulation of motor behavior by DA neurons within multiple hypothalamic nuclei and elucidate a novel functional mechanism for the preoptic DA neurons in the initiation of movement.
Collapse
Affiliation(s)
- Joshua P Barrios
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Wei-Chun Wang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Roman England
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Erica Reifenberg
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam D Douglass
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
13
|
Son JH, Stevenson TJ, Bowles MD, Scholl EA, Bonkowsky JL. Dopaminergic Co-Regulation of Locomotor Development and Motor Neuron Synaptogenesis is Uncoupled by Hypoxia in Zebrafish. eNeuro 2020; 7:ENEURO.0355-19.2020. [PMID: 32001551 PMCID: PMC7046933 DOI: 10.1523/eneuro.0355-19.2020] [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: 08/26/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 11/21/2022] Open
Abstract
Hypoxic injury to the developing human brain is a complication of premature birth and is associated with long-term impairments of motor function. Disruptions of axon and synaptic connectivity have been linked to developmental hypoxia, but the fundamental mechanisms impacting motor function from altered connectivity are poorly understood. We investigated the effects of hypoxia on locomotor development in zebrafish. We found that developmental hypoxia resulted in decreased spontaneous swimming behavior in larva, and that this motor impairment persisted into adulthood. In evaluation of the diencephalic dopaminergic neurons, which regulate early development of locomotion and constitute an evolutionarily conserved component of the vertebrate dopaminergic system, hypoxia caused a decrease in the number of synapses from the descending dopaminergic diencephalospinal tract (DDT) to spinal cord motor neurons. Moreover, dopamine signaling from the DDT was coupled jointly to motor neuron synaptogenesis and to locomotor development. Together, these results demonstrate the developmental processes regulating early locomotor development and a requirement for dopaminergic projections and motor neuron synaptogenesis. Our findings suggest new insights for understanding the mechanisms leading to motor disability from hypoxic injury of prematurity.
Collapse
Affiliation(s)
- Jong-Hyun Son
- Department of Biology, University of Scranton, Scranton, PA 18510
| | - Tamara J Stevenson
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Miranda D Bowles
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Erika A Scholl
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84132
- Brain and Spine Center, Primary Children's Hospital, Salt Lake City, UT 84108
| |
Collapse
|
14
|
Jha U, Thirumalai V. Neuromodulatory Selection of Motor Neuron Recruitment Patterns in a Visuomotor Behavior Increases Speed. Curr Biol 2020; 30:788-801.e3. [PMID: 32084402 DOI: 10.1016/j.cub.2019.12.064] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/18/2019] [Accepted: 12/19/2019] [Indexed: 01/26/2023]
Abstract
Animals generate locomotion at different speeds to suit their behavioral needs. Spinal circuits generate locomotion at these varying speeds by sequential activation of different spinal interneurons and motor neurons. Larval zebrafish can generate slow swims for prey capture and exploration by activation of secondary motor neurons and much faster and vigorous swims during escape and struggle via additional activation of primary motor neurons. Neuromodulators are known to alter the motor output of spinal circuits, but their precise role in speed regulation is not well understood. Here, in the context of optomotor response (OMR), an innate evoked locomotor behavior, we show that dopamine (DA) provides an additional layer to regulation of swim speed in larval zebrafish. Activation of D1-like receptors increases swim speed during OMR in free-swimming larvae. By analyzing tail bend kinematics in head-restrained larvae, we show that the increase in speed is actuated by larger tail bends. Whole-cell patch-clamp recordings from motor neurons reveal that, during OMR, typically only secondary motor neurons are active, whereas primary motor neurons are quiescent. Activation of D1-like receptors increases intrinsic excitability and excitatory synaptic drive in primary and secondary motor neurons. These actions result in greater recruitment of motor neurons during OMR. Our findings provide an example of neuromodulatory reconfiguration of spinal motor neuron speed modules where members are selectively recruited and motor drive is increased to effect changes in locomotor speed. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Urvashi Jha
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India; SASTRA Deemed University, School of Chemical and Biotechnology, Thanjavur, Tamil Nadu 613401, India
| | - Vatsala Thirumalai
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India.
| |
Collapse
|
15
|
JavadiEsfahani R, Kwong RWM. The sensory-motor responses to environmental acidosis in larval zebrafish: Influences of neurotransmitter and water chemistry. CHEMOSPHERE 2019; 235:383-390. [PMID: 31271998 DOI: 10.1016/j.chemosphere.2019.06.133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 06/09/2023]
Abstract
The sensory-motor function in larval zebrafish (Danio rerio) following exposure to low water pH was investigated. The results suggested that acid exposure (pH 4.0-5.0; control: pH 7.4) significantly reduced the touch-evoked escape response of larval zebrafish at 3 days post fertilization (dpf). A significant number of pH 4.0-exposed larvae also exhibited a lack of escape response. Treatment with neurotransmitters showed that serotonin or acetylcholine, but not dopamine, reduced the adverse effects of acid exposure on the escape response of larvae. Co-exposure to serotonin and acetylcholine did not further improve the escape response of acid-exposed larvae, suggesting no additive effect by these neurotransmitters. Interestingly, the negative effects of acid exposure on the escape response could be completely rescued by elevating the water levels of Ca2+, but not NaCl. Collectively, these results suggested that acid-induced disruption in Ca2+ balance suppressed the serotonin- and acetylcholine-mediated neuronal signaling, thereby affecting the sensory-motor function and escape response of larval zebrafish. Findings from the present study may have important implication for the survival (e.g., escape from adverse conditions) of larval fish in acid-impacted environments, particularly during early development when they are still incapable of spontaneous swimming.
Collapse
|
16
|
Abstract
Visual stimuli can evoke complex behavioral responses, but the underlying streams of neural activity in mammalian brains are difficult to follow because of their size. Here, I review the visual system of zebrafish larvae, highlighting where recent experimental evidence has localized the functional steps of visuomotor transformations to specific brain areas. The retina of a larva encodes behaviorally relevant visual information in neural activity distributed across feature-selective ganglion cells such that signals representing distinct stimulus properties arrive in different areas or layers of the brain. Motor centers in the hindbrain encode motor variables that are precisely tuned to behavioral needs within a given stimulus setting. Owing to rapid technological progress, larval zebrafish provide unique opportunities for obtaining a comprehensive understanding of the intermediate processing steps occurring between visual and motor centers, revealing how visuomotor transformations are implemented in a vertebrate brain.
Collapse
Affiliation(s)
- Johann H. Bollmann
- Developmental Biology, Institute of Biology I, Faculty of Biology, and Bernstein Center Freiburg, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
17
|
Kalichak F, de Alcantara Barcellos HH, Idalencio R, Koakoski G, Soares SM, Pompermaier A, Rossini M, Barcellos LJG. Persistent and transgenerational effects of risperidone in zebrafish. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:26293-26303. [PMID: 31286368 DOI: 10.1007/s11356-019-05890-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 07/01/2019] [Indexed: 06/09/2023]
Abstract
Since behavior is the connection between the internal physiological processes of an animal and its interaction with the environment, a complete behavioral repertoire is crucial for fish survival and fitness, at both the individual and population levels. Thus, unintended exposure of non-target organisms to antipsychotic residues in the environment can impact their normal behavior, and some of these behavioral changes can be seen during the entire life of the animal and passed to subsequent generations. Although there are some reports related to transgenerational toxicology, little is known of the long-term consequences of exposure to pharmaceutical compounds such as risperidone. Here, we show that zebrafish exposed to risperidone (RISP) during embryonic and larval stages presented impaired anti-predatory behavior during adulthood, characterizing a persistent effect. We also show that some of these behavioral changes are present in the following generation, characterizing a transgenerational effect. This suggests that even short exposures to environmentally relevant concentrations, at essential stages of development, can persist throughout the whole life of the zebrafish, including its offspring. From an environmental perspective, our results suggested possible risks and long-term consequences associated with drug residues in water, which can affect aquatic life and endanger species that depend on appropriate behavioral responses for survival.
Collapse
Affiliation(s)
- Fabiana Kalichak
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Curso de Medicina Veterinária, Faculdades Integradas do Vale do Iguaçu (Uniguaçu), Rua Padre Saporiti, 717, Rio D'Areia, União da Vitória, PR, 84600-904, Brazil
| | - Heloisa Helena de Alcantara Barcellos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Curso de Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Renan Idalencio
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Curso de Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Gessi Koakoski
- Programa de Pós-Graduação em Bioexperimentação, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Suelen Mendonça Soares
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Aline Pompermaier
- Programa de Pós-Graduação em Ciências Ambientais, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Mainara Rossini
- Curso de Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Leonardo José Gil Barcellos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil.
- Curso de Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil.
- Programa de Pós-Graduação em Bioexperimentação, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil.
- Programa de Pós-Graduação em Ciências Ambientais, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil.
| |
Collapse
|
18
|
Regeneration of Dopaminergic Neurons in Adult Zebrafish Depends on Immune System Activation and Differs for Distinct Populations. J Neurosci 2019; 39:4694-4713. [PMID: 30948475 DOI: 10.1523/jneurosci.2706-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/23/2022] Open
Abstract
Adult zebrafish, in contrast to mammals, regenerate neurons in their brain, but the extent and variability of this capacity is unclear. Here we ask whether the loss of various dopaminergic neuron populations is sufficient to trigger their functional regeneration. Both sexes of zebrafish were analyzed. Genetic lineage tracing shows that specific diencephalic ependymo-radial glial (ERG) progenitor cells give rise to new dopaminergic [tyrosine hydroxylase-positive (TH+)] neurons. Ablation elicits an immune response, increased proliferation of ERG progenitor cells, and increased addition of new TH+ neurons in populations that constitutively add new neurons (e.g., diencephalic population 5/6). Inhibiting the immune response attenuates neurogenesis to control levels. Boosting the immune response enhances ERG proliferation, but not addition of TH+ neurons. In contrast, in populations in which constitutive neurogenesis is undetectable (e.g., the posterior tuberculum and locus ceruleus), cell replacement and tissue integration are incomplete and transient. This is associated with a loss of spinal TH+ axons, as well as permanent deficits in shoaling and reproductive behavior. Hence, dopaminergic neuron populations in the adult zebrafish brain show vast differences in regenerative capacity that correlate with constitutive addition of neurons and depend on immune system activation.SIGNIFICANCE STATEMENT Despite the fact that zebrafish show a high propensity to regenerate neurons in the brain, this study reveals that not all types of dopaminergic neurons are functionally regenerated after specific ablation. Hence, in the same adult vertebrate brain, mechanisms of successful and incomplete regeneration can be studied. We identify progenitor cells for dopaminergic neurons and show that activating the immune system promotes the proliferation of these cells. However, in some areas of the brain this only leads to insufficient replacement of functionally important dopaminergic neurons that later disappear. Understanding the mechanisms of regeneration in zebrafish may inform interventions targeting the regeneration of functionally important neurons, such as dopaminergic neurons, from endogenous progenitor cells in nonregenerating mammals.
Collapse
|
19
|
do Carmo Silva RX, Lima-Maximino MG, Maximino C. The aversive brain system of teleosts: Implications for neuroscience and biological psychiatry. Neurosci Biobehav Rev 2018; 95:123-135. [DOI: 10.1016/j.neubiorev.2018.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/24/2022]
|
20
|
Prenatal Neuropathologies in Autism Spectrum Disorder and Intellectual Disability: The Gestation of a Comprehensive Zebrafish Model. J Dev Biol 2018; 6:jdb6040029. [PMID: 30513623 PMCID: PMC6316217 DOI: 10.3390/jdb6040029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) and intellectual disability (ID) are neurodevelopmental disorders with overlapping diagnostic behaviors and risk factors. These include embryonic exposure to teratogens and mutations in genes that have important functions prenatally. Animal models, including rodents and zebrafish, have been essential in delineating mechanisms of neuropathology and identifying developmental critical periods, when those mechanisms are most sensitive to disruption. This review focuses on how the developmentally accessible zebrafish is contributing to our understanding of prenatal pathologies that set the stage for later ASD-ID behavioral deficits. We discuss the known factors that contribute prenatally to ASD-ID and the recent use of zebrafish to model deficits in brain morphogenesis and circuit development. We conclude by suggesting that a future challenge in zebrafish ASD-ID modeling will be to bridge prenatal anatomical and physiological pathologies to behavioral deficits later in life.
Collapse
|
21
|
Calienni MN, Temprana CF, Prieto MJ, Paolino D, Fresta M, Tekinay AB, Alonso SDV, Montanari J. Nano-formulation for topical treatment of precancerous lesions: skin penetration, in vitro, and in vivo toxicological evaluation. Drug Deliv Transl Res 2018; 8:496-514. [PMID: 29288359 DOI: 10.1007/s13346-017-0469-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
With the aim of improving the topical delivery of the antineoplastic drug 5-fluorouracil (5FU), it was loaded into ultradeformable liposomes composed of soy phosphatidylcholine and sodium cholate (UDL-5FU). The liposome populations had a mean size of 70 nm without significant changes in 56 days, and the ultradeformable formulations were up to 324-fold more elastic than conventional liposomes. The interaction between 5FU and the liposomal membrane was studied by three methods, and also release profile was obtained. UDL-5FU did penetrate the stratum corneum of human skin. At in vitro experiments, the formulation was more toxic on a human melanoma-derived than on a human keratinocyte-derived cell line. Cells captured liposomes by metabolically active processes. In vivo toxicity experiments were carried out in zebrafish (Danio rerio) larvae by studying the swimming activity, morphological changes, and alterations in the heart rate after incubation. UDL-5FU was more toxic than free 5FU. Therefore, this nano-formulation could be useful for topical application in deep skin precancerous lesions with advantages over current treatments. This is the first work that assessed the induction of apoptosis, skin penetration in a Saarbrücken penetration model, and the toxicological effects in vivo of an ultradeformable 5FU-loaded formulation.
Collapse
Affiliation(s)
- Maria Natalia Calienni
- Laboratorio de Biomembranas, Departamento de Ciencia y Tecnología, GBEyB. IMBICE, CCT-LA PLATA, CONICET, Universidad Nacional de Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina.,Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Carlos Facundo Temprana
- Laboratorio de Inmunología y Virología (LIV), Departamento de Ciencia y Tecnología, CONICET, Universidad Nacional de Quilmes, B1876BXD, Bernal, Argentina
| | - Maria Jimena Prieto
- Laboratorio de Biomembranas, Departamento de Ciencia y Tecnología, GBEyB. IMBICE, CCT-LA PLATA, CONICET, Universidad Nacional de Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Massimo Fresta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Viale S. Venuta, Germaneto, I-88100, Catanzaro, Italy
| | - Ayse Begum Tekinay
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, 06800, Ankara, Turkey
| | - Silvia Del Valle Alonso
- Laboratorio de Biomembranas, Departamento de Ciencia y Tecnología, GBEyB. IMBICE, CCT-LA PLATA, CONICET, Universidad Nacional de Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| | - Jorge Montanari
- Laboratorio de Biomembranas, Departamento de Ciencia y Tecnología, GBEyB. IMBICE, CCT-LA PLATA, CONICET, Universidad Nacional de Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina. .,Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, 06800, Ankara, Turkey.
| |
Collapse
|
22
|
Clements KN, Miller TH, Keever JM, Hall AM, Issa FA. Social Status-Related Differences in Motor Activity Between Wild-Type and Mutant Zebrafish. THE BIOLOGICAL BULLETIN 2018; 235:71-82. [PMID: 30358446 DOI: 10.1086/699514] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Use of zebrafish as a model organism in biomedical research has led to the generation of many genetically modified mutant lines to investigate various aspects of developmental and cellular processes. However, the broader effects of the underlying mutations on social and motor behavior remain poorly examined. Here, we compared the dynamics of social interactions in the Tüpfel long-fin nacre mutant line, which lacks skin pigmentation, to wild-type zebrafish; and we determined whether status-dependent differences in escape and swimming behavior existed within each strain. We show that despite similarities in aggressive activity, Tüpfel long-fin nacre pairs exhibit unstable social relationships characterized by frequent reversals in social dominance compared to wild-type pairs. The lack of strong dominance relationships in Tüpfel long-fin nacre pairs correlates with weak territoriality and overlapping spatial distribution of dominants and subordinates. Conversely, wild-type dominants displayed strong territoriality that severely limited the movement of subordinates. Additionally, the sensitivity of the startle escape response was significantly higher in wild-type subordinates compared to dominants. However, status-related differences in sensitivity of escape response in Tüpfel long-fin nacre pairs were absent. Finally, we present evidence suggesting that these differences could be a consequence of a disruption of proper visual social signals. We show that in wild-type pairs dominants are more conspicuous, and that in wild-type and Tüpfel long-fin nacre pairings wild-type fish are more likely to dominate Tüpfel long-fin nacres. Our results serve as a cautionary note in research design when morphologically engineered zebrafish for color differences are utilized in the study of social behavior and central nervous system function.
Collapse
|
23
|
Berg EM, Björnfors ER, Pallucchi I, Picton LD, El Manira A. Principles Governing Locomotion in Vertebrates: Lessons From Zebrafish. Front Neural Circuits 2018; 12:73. [PMID: 30271327 PMCID: PMC6146226 DOI: 10.3389/fncir.2018.00073] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/27/2018] [Indexed: 11/24/2022] Open
Abstract
Locomotor behaviors are critical for survival and enable animals to navigate their environment, find food and evade predators. The circuits in the brain and spinal cord that initiate and maintain such different modes of locomotion in vertebrates have been studied in numerous species for over a century. In recent decades, the zebrafish has emerged as one of the main model systems for the study of locomotion, owing to its experimental amenability, and work in zebrafish has revealed numerous new insights into locomotor circuit function. Here, we review the literature that has led to our current understanding of the neural circuits controlling swimming and escape in zebrafish. We highlight recent studies that have enriched our comprehension of key topics, such as the interactions between premotor excitatory interneurons (INs) and motoneurons (MNs), supraspinal and spinal circuits that coordinate escape maneuvers, and developmental changes in overall circuit composition. We also discuss roles for neuromodulators and sensory inputs in modifying the relative strengths of constituent circuit components to provide flexibility in zebrafish behavior, allowing the animal to accommodate changes in the environment. We aim to provide a coherent framework for understanding the circuitry in the brain and spinal cord of zebrafish that allows the animal to flexibly transition between different speeds, and modes, of locomotion.
Collapse
Affiliation(s)
- Eva M Berg
- Department of Neuroscience, Karolinska Institute (KI), Stockholm, Sweden
| | | | - Irene Pallucchi
- Department of Neuroscience, Karolinska Institute (KI), Stockholm, Sweden
| | - Laurence D Picton
- Department of Neuroscience, Karolinska Institute (KI), Stockholm, Sweden
| | | |
Collapse
|
24
|
Lobao-Soares B, Eduardo-da-Silva P, Amarilha H, Pinheiro-da-Silva J, Silva PF, Luchiari AC. It's Tea Time: Interference of Ayahuasca Brew on Discriminative Learning in Zebrafish. Front Behav Neurosci 2018; 12:190. [PMID: 30210319 PMCID: PMC6119691 DOI: 10.3389/fnbeh.2018.00190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/06/2018] [Indexed: 12/01/2022] Open
Abstract
Ayahuasca is a psychoactive brew traditionally used in shamanistic and vegetalistic rituals and has recently received lot of attention due to potential cognitive benefits. Ayahuasca effects are caused by the synergistic interaction of β-carbolines (harmine, harmaline and tetrahydroarmine) contained in Banisteriopsis caapi stalks combined with the N,N-dimethyltryptamine (DMT) from Psychotria viridis leaves, a potent agonist to serotonin (5-HT) receptors. The present study approaches the effects of chronic and acute exposure to two Ayahuasca concentrations (0.1 and 0.5 ml/L) on the cognitive ability to discriminate objects in a one-trial learning task in zebrafish. Based on the combination of concentrations and exposure regimens, we divided adult zebrafish in five treatment groups: acute 0.1 and 0.5 ml/L, chronic 0.1 and 0.5 ml/L, and control 0.0 (n = 20 for each group). Then we tested them in a memory task of object discrimination. Acute Ayahuasca exposed groups performed similarly to the control group, however chronically treated fish (13 days) presented both impaired discriminative performance and locomotor alterations. Overall, these results indicate that Ayahuasca is a potent psychoactive drug that, in chronic exposure, negatively affects mnemonic parameters in zebrafish. In single exposure it does not affects cognitive performance, but the higher concentration (0.5) affected locomotion. Moreover, we reinforce the importance of the zebrafish for behavioral pharmacological studies of drug screening, in special to psychedelic drug research.
Collapse
Affiliation(s)
- Bruno Lobao-Soares
- Departamento de Biofísica e Farmacologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Paulianny Eduardo-da-Silva
- Departamento de Biofísica e Farmacologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Hugo Amarilha
- Departamento de Biofísica e Farmacologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | | | - Priscila F. Silva
- Departamento de Fisiologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Ana Carolina Luchiari
- Departamento de Fisiologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
25
|
Robinson B, Dumas M, Gu Q, Kanungo J. N-acetylcysteine prevents ketamine-induced adverse effects on development, heart rate and monoaminergic neurons in zebrafish. Neurosci Lett 2018; 682:56-61. [PMID: 29890257 PMCID: PMC6102060 DOI: 10.1016/j.neulet.2018.06.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 01/11/2023]
Abstract
N-acetylcysteine, a precursor molecule of glutathione, is an antioxidant. Ketamine, a pediatric anesthetic, has been implicated in cardiotoxicity and neurotoxicity including modulation of monoaminergic systems in mammals and zebrafish. Here, we show that N-acetylcysteine prevents ketamine's adverse effects on development and monoaminergic neurons in zebrafish embryos. The effects of ketamine and N-acetylcysteine alone or in combination were measured on the heart rate, body length, brain serotonergic neurons and tyrosine hydroxylase-immunoreactive (TH-IR) neurons. In the absence of N-acetylcysteine, a concentration of ketamine that produces an internal embryo exposure level comparable to human anesthetic plasma concentrations significantly reduced heart rate and body length and those effects were prevented by N-acetylcysteine co-treatment. Ketamine also reduced the areas occupied by serotonergic neurons in the brain, whereas N-acetylcysteine co-exposure counteracted this effect. TH-IR neurons in the embryo brain and TH-IR cells in the trunk were significantly reduced with ketamine treatment, but not in the presence of N-acetylcysteine. In our continued search for compounds that can prevent ketamine toxicity, this study using specific endpoints of developmental toxicity, cardiotoxicity and neurotoxicity, demonstrates protective effects of N-acetylcysteine against ketamine's adverse effects. This is the first study that shows the protective effects of N-acetylcysteine on ketamine-induced developmental defects of monoaminergic neurons as observed in a whole organism.
Collapse
Affiliation(s)
- Bonnie Robinson
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Melanie Dumas
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Qiang Gu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| |
Collapse
|
26
|
Neural Cotransmission in Spinal Circuits Governing Locomotion. Trends Neurosci 2018; 41:540-550. [DOI: 10.1016/j.tins.2018.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/06/2018] [Accepted: 04/17/2018] [Indexed: 01/08/2023]
|
27
|
Montgomery JE, Wahlstrom-Helgren S, Wiggin TD, Corwin BM, Lillesaar C, Masino MA. Intraspinal serotonergic signaling suppresses locomotor activity in larval zebrafish. Dev Neurobiol 2018; 78:10.1002/dneu.22606. [PMID: 29923318 PMCID: PMC6301152 DOI: 10.1002/dneu.22606] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/27/2018] [Accepted: 05/17/2018] [Indexed: 12/27/2022]
Abstract
Serotonin (5HT) is a modulator of many vital processes in the spinal cord (SC), such as production of locomotion. In the larval zebrafish, intraspinal serotonergic neurons (ISNs) are a source of spinal 5HT that, despite the availability of numerous genetic and optical tools, has not yet been directly shown to affect the spinal locomotor network. In order to better understand the functions of ISNs, we used a combination of strategies to investigate ISN development, morphology, and function. ISNs were optically isolated from one another by photoconverting Kaede fluorescent protein in individual cells, permitting morphometric analysis as they developed in vivo. ISN neurite lengths and projection distances exhibited the greatest amount of change between 3 and 4 days post-fertilization (dpf) and appeared to stabilize by 5 dpf. Overall ISN innervation patterns were similar between cells and between SC regions. ISNs possessed rostrally-extending neurites resembling dendrites and a caudally-extending neurite resembling an axon, which terminated with an enlarged growth cone-like structure. Interestingly, these enlargements remained even after neurite extension had ceased. Functionally, application of exogenous 5HT reduced spinally-produced motor nerve bursting. A selective 5HT reuptake inhibitor and ISN activation with channelrhodopsin-2 each produced similar effects to 5HT, indicating that spinally-intrinsic 5HT originating from the ISNs has an inhibitory effect on the spinal locomotor network. Taken together this suggests that the ISNs are morphologically mature by 5 dpf and supports their involvement in modulating the activity of the spinal locomotor network. © 2018 Wiley Periodicals, Inc. Develop Neurobiol, 2018.
Collapse
Affiliation(s)
| | | | - Timothy D. Wiggin
- University of Minnesota, Department of Neuroscience, Minneapolis, MN
| | | | - Christina Lillesaar
- University of Würzburg, Department of Physiological Chemistry, Biocenter, Würzburg, Germany
| | - Mark A. Masino
- University of Minnesota, Department of Neuroscience, Minneapolis, MN
| |
Collapse
|
28
|
Park C, Clements KN, Issa FA, Ahn S. Effects of Social Experience on the Habituation Rate of Zebrafish Startle Escape Response: Empirical and Computational Analyses. Front Neural Circuits 2018; 12:7. [PMID: 29459823 PMCID: PMC5807392 DOI: 10.3389/fncir.2018.00007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
While the effects of social experience on nervous system function have been extensively investigated in both vertebrate and invertebrate systems, our understanding of how social status differentially affects learning remains limited. In the context of habituation, a well-characterized form of non-associative learning, we investigated how the learning processes differ between socially dominant and subordinate in zebrafish (Danio rerio). We found that social status and frequency of stimulus inputs influence the habituation rate of short latency C-start escape response that is initiated by the Mauthner neuron (M-cell). Socially dominant animals exhibited higher habituation rates compared to socially subordinate animals at a moderate stimulus frequency, but low stimulus frequency eliminated this difference of habituation rates between the two social phenotypes. Moreover, habituation rates of both dominants and subordinates were higher at a moderate stimulus frequency compared to those at a low stimulus frequency. We investigated a potential mechanism underlying these status-dependent differences by constructing a simplified neurocomputational model of the M-cell escape circuit. The computational study showed that the change in total net excitability of the model M-cell was able to replicate the experimental results. At moderate stimulus frequency, the model M-cell with lower total net excitability, that mimicked a dominant-like phenotype, exhibited higher habituation rates. On the other hand, the model with higher total net excitability, that mimicked the subordinate-like phenotype, exhibited lower habituation rates. The relationship between habituation rates and characteristics (frequency and amplitude) of the repeated stimulus were also investigated. We found that habituation rates are decreasing functions of amplitude and increasing functions of frequency while these rates depend on social status (higher for dominants and lower for subordinates). Our results show that social status affects habituative learning in zebrafish, which could be mediated by a summative neuromodulatory input to the M-cell escape circuit, which enables animals to readily learn to adapt to changes in their social environment.
Collapse
Affiliation(s)
- Choongseok Park
- Department of Mathematics, North Carolina A&T State University, Greensboro, NC, United States
| | - Katie N Clements
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Fadi A Issa
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Sungwoo Ahn
- Department of Mathematics, East Carolina University, Greenville, NC, United States
| |
Collapse
|
29
|
Calienni MN, Feas DA, Igartúa DE, Chiaramoni NS, Alonso SDV, Prieto MJ. Nanotoxicological and teratogenic effects: A linkage between dendrimer surface charge and zebrafish developmental stages. Toxicol Appl Pharmacol 2017; 337:1-11. [DOI: 10.1016/j.taap.2017.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 01/02/2023]
|
30
|
Kalichak F, Idalencio R, da Rosa JGS, Barcellos HHDA, Fagundes M, Piato A, Barcellos LJG. Psychotropic in the environment: risperidone residues affect the behavior of fish larvae. Sci Rep 2017; 7:14121. [PMID: 29074994 PMCID: PMC5658348 DOI: 10.1038/s41598-017-14575-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/04/2017] [Indexed: 11/08/2022] Open
Abstract
The ability to avoid and escape from predators are clearly relevant behaviors from the ecological perspective and directly interfere with the survival of organisms. Detected in the aquatic environment, risperidone can alter the behavior of exposed species. Considering the risk of exposure in the early stages of life, we exposed zebrafish embryos to risperidone during the first 5 days of life. Risperidone caused hyperactivity in exposed larvae, which in an environmental context, the animals may be more vulnerable to predation due to greater visibility or less perception of risk areas.
Collapse
Affiliation(s)
- Fabiana Kalichak
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Renan Idalencio
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - João Gabriel Santos da Rosa
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Heloísa Helena de Alcântara Barcellos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Michele Fagundes
- Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
- Programa de Pós-Graduação em Ciências Ambientais, Instituto de Ciências Biológicas, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Angelo Piato
- Programa de Pós-Graduação em Farmacologia e Terapêutica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Sarmento Leite 500/305, Porto Alegre, RS, 90050-170, Brazil
| | - Leonardo José Gil Barcellos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil.
- Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil.
- Programa de Pós-Graduação em Ciências Ambientais, Instituto de Ciências Biológicas, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil.
- Programa de Pós-Graduação em Bioexperimentação, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil.
| |
Collapse
|
31
|
Zebrafish slc30a10 deficiency revealed a novel compensatory mechanism of Atp2c1 in maintaining manganese homeostasis. PLoS Genet 2017; 13:e1006892. [PMID: 28692648 PMCID: PMC5524415 DOI: 10.1371/journal.pgen.1006892] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/24/2017] [Accepted: 06/23/2017] [Indexed: 12/28/2022] Open
Abstract
Recent studies found that mutations in the human SLC30A10 gene, which encodes a manganese (Mn) efflux transporter, are associated with hypermanganesemia with dystonia, polycythemia, and cirrhosis (HMDPC). However, the relationship between Mn metabolism and HMDPC is poorly understood, and no specific treatments are available for this disorder. Here, we generated two zebrafish slc30a10 mutant lines using the CRISPR/Cas9 system. Compared to wild-type animals, mutant adult animals developed significantly higher systemic Mn levels, and Mn accumulated in the brain and liver of mutant embryos in response to exogenous Mn. Interestingly, slc30a10 mutants developed neurological deficits in adulthood, as well as environmental Mn-induced manganism in the embryonic stage; moreover, mutant animals had impaired dopaminergic and GABAergic signaling. Finally, mutant animals developed steatosis, liver fibrosis, and polycythemia accompanied by increased epo expression. This phenotype was rescued partially by EDTA- CaNa2 chelation therapy and iron supplementation. Interestingly, prior to the onset of slc30a10 expression, expressing ATP2C1 (ATPase secretory pathway Ca2+ transporting 1) protected mutant embryos from Mn exposure, suggesting a compensatory role for Atp2c1 in the absence of Slc30a10. Notably, expressing either wild-type or mutant forms of SLC30A10 was sufficient to inhibit the effect of ATP2C1 in response to Mn challenge in both zebrafish embryos and HeLa cells. These findings suggest that either activating ATP2C1 or restoring the Mn-induced trafficking of ATP2C1 can reduce Mn accumulation, providing a possible target for treating HMDPC. Impaired function of the manganese transporter SLC30A10 has been implicated in HMDPC (hypermanganesemia with dystonia, polycythemia, and cirrhosis), an early-onset metabolic disorder clinically characterized by increased systemic Mn levels, neurological impairment, polycythemia, and hepatic injury. No specific treatment is currently available for HMDPC. Moreover, the mechanisms that underlie Mn metabolism are poorly understood, thereby hindering the development of effective treatments. To investigate the physiological processes underlying Mn metabolism and to develop new disease models of HMDPC, we generated two zebrafish slc30a10 mutant lines using the CRISPR/Cas9 system and found that these mutants develop clinical deficits typically associated with HMDPC. Furthermore, we identified a putative compensatory role for ATP2C1 in the absence of SLC30A10 with respect to modulating Mn metabolism. These findings provide a valuable tool for investigating the role of manganese dysregulation in neurological degenerative diseases and which can be used to develop new pharmacological approaches for managing Mn accumulation.
Collapse
|
32
|
Storage of neural histamine and histaminergic neurotransmission is VMAT2 dependent in the zebrafish. Sci Rep 2017; 7:3060. [PMID: 28596586 PMCID: PMC5465064 DOI: 10.1038/s41598-017-02981-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/20/2017] [Indexed: 11/09/2022] Open
Abstract
Monoaminergic neurotransmission is greatly dependent on the function of the vesicular monoamine transporter VMAT2, which is responsible for loading monoamines into secretory vesicles. The role of VMAT2 in histaminergic neurotransmission is poorly understood. We studied the structure and function of the histaminergic system in larval zebrafish following inhibition of VMAT2 function by reserpine. We found that reserpine treatment greatly reduced histamine immunoreactivity in neurons and an almost total disappearance of histamine-containing nerve fibers in the dorsal telencephalon and habenula, the most densely innervated targets of the hypothalamic histamine neurons. The reserpine treated larvae had an impaired histamine-dependent dark-induced flash response seen during the first second after onset of darkness, implying that function of the histaminergic network is VMAT2 dependent. Levels of histamine and other monoamines were decreased in reserpine treated animals. This study provides conclusive evidence of the relevance of VMAT2 in histaminergic neurotransmission, further implying that the storage and release mechanism of neural histamine is comparable to that of other monoamines. Our results also reveal potential new insights about the roles of monoaminergic neurotransmitters in the regulation of locomotion increase during adaptation to darkness.
Collapse
|
33
|
Huang SB, Zhao HD, Wang LF, Sun MF, Zhu YL, Wu YB, Xu YD, Peng SX, Cui C, Shen YQ. Intradiencephalon injection of histamine inhibited the recovery of locomotor function of spinal cord injured zebrafish. Biochem Biophys Res Commun 2017; 489:275-280. [PMID: 28559136 DOI: 10.1016/j.bbrc.2017.05.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 01/22/2023]
Abstract
Human spinal cord injury (SCI) usually causes irreversible disability beneath the injured site due to poor neural regeneration. On the contrary, zebrafish show significant regenerative ability after SCI, thus is usually worked as an animal model for studying neuroregeneration. Most of the previous SCI studies focused on the local site of SCI, the supraspinal-derived signals were rarely mentioned. Here we showed that intradiencephalon injection of histamine (HA) inhibited the locomotor recovery in adult zebrafish post-SCI. Immunofluorescence results showed that intradiencephalon HA administration increased the activated microglia 3 days post injury (dpi), promoted the proliferation of radial glial cells at 7 dpi and affected the morphology of radial glial cells at 11 dpi. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) results showed that intradiencephalon HA administration also reduced the expression of neurotrophic factors including brain-derived neurotrophic factor (BDNF) and insulin-like growth factor1 (IGF-1) at the lesion site, however, had no effect on the expression of pro-inflammatory factors such as TNF-alpha and IL-1 beta. Hence, our data suggested that exogenous intradiencephalon HA retarded locomotor recovery in spinal cord injured zebrafish via modulating the repair microenvironment.
Collapse
Affiliation(s)
- Shu-Bing Huang
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Hou-De Zhao
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Lin-Fang Wang
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Meng-Fei Sun
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Ying-Li Zhu
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Yi-Bo Wu
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China; Affiliated Hospital of Jiangnan University, Human Reproductive and Genetic Center, Wuxi, China
| | - Yi-Da Xu
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Shi-Xiao Peng
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China
| | - Chun Cui
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China.
| | - Yan-Qin Shen
- Neuroscience Center, Jiangnan University Medical School, Wuxi, China.
| |
Collapse
|
34
|
Social Status-Dependent Shift in Neural Circuit Activation Affects Decision Making. J Neurosci 2017; 37:2137-2148. [PMID: 28093472 DOI: 10.1523/jneurosci.1548-16.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 11/21/2022] Open
Abstract
In a social group, animals make behavioral decisions that fit their social ranks. These behavioral choices are dependent on the various social cues experienced during social interactions. In vertebrates, little is known of how social status affects the underlying neural mechanisms regulating decision-making circuits that drive competing behaviors. Here, we demonstrate that social status in zebrafish (Danio rerio) influences behavioral decisions by shifting the balance in neural circuit activation between two competing networks (escape and swim). We show that socially dominant animals enhance activation of the swim circuit. Conversely, social subordinates display a decreased activation of the swim circuit, but an enhanced activation of the escape circuit. In an effort to understand how social status mediates these effects, we constructed a neurocomputational model of the escape and swim circuits. The model replicates our findings and suggests that social status-related shift in circuit dynamics could be mediated by changes in the relative excitability of the escape and swim networks. Together, our results reveal that changes in the excitabilities of the Mauthner command neuron for escape and the inhibitory interneurons that regulate swimming provide a cellular mechanism for the nervous system to adapt to changes in social conditions by permitting the animal to select a socially appropriate behavioral response.SIGNIFICANCE STATEMENT Understanding how social factors influence nervous system function is of great importance. Using zebrafish as a model system, we demonstrate how social experience affects decision making to enable animals to produce socially appropriate behavior. Based on experimental evidence and computational modeling, we show that behavioral decisions reflect the interplay between competing neural circuits whose activation thresholds shift in accordance with social status. We demonstrate this through analysis of the behavior and neural circuit responses that drive escape and swim behaviors in fish. We show that socially subordinate animals favor escape over swimming, while socially dominants favor swimming over escape. We propose that these differences are mediated by shifts in relative circuit excitability.
Collapse
|
35
|
The Descending Diencephalic Dopamine System Is Tuned to Sensory Stimuli. Curr Biol 2017; 27:318-333. [PMID: 28089511 DOI: 10.1016/j.cub.2016.11.059] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 10/31/2016] [Accepted: 11/30/2016] [Indexed: 12/22/2022]
Abstract
The vertebrate diencephalic A11 system provides the sole dopaminergic innervation of hindbrain and spinal cord and has been implicated in modulation of locomotion and sensory processes. However, the exact contributions of sensory stimuli and motor behavior to A11 dopaminergic activity remain unclear. We recorded cellular calcium activity in four anatomically distinct posterior tubercular A11-type dopaminergic subgroups and two adjacent hypothalamic dopaminergic groups in GCaMP7a-transgenic, semi-restrained zebrafish larvae. Our analyses reveal the contributions of different sensory modalities and motor states to dopaminergic activity. Each posterior tubercular and hypothalamic subgroup showed distinct activity patterns, while activity was synchronous within individual subgroups. Caudal and dorsomedial hypothalamic dopaminergic neurons are activated following vigorous tail movements and stay active for about 10 s, revealing predominantly post-motor activity. In contrast, posterior tubercular dopaminergic neurons are predominantly sensory driven, with subgroups differentially responding to different tactile or visual sensory modalities. In the anterior subgroups, neuronal response magnitudes are tuned to tactile stimulus intensities, revealing features similar to sensory systems. We identify the lateral line system as source for this tactile tuning. In contrast, the posterior subgroup is responsive to distinct moving visual stimuli. Specifically, translational forward stimuli, which may indicate insufficient rheotaxis and drift, induce dopaminergic activity, but backward or rotational stimuli not. The activation of posterior tubercular dopaminergic neurons by sensory stimuli, and their projections onto peripheral mechanosensory systems, suggests a participation of A11-type neurons in the feedback regulation of sensory systems. Together with the adjacent hypothalamic neurons, they may serve to set basic behavioral states.
Collapse
|
36
|
Kawashima T, Zwart MF, Yang CT, Mensh BD, Ahrens MB. The Serotonergic System Tracks the Outcomes of Actions to Mediate Short-Term Motor Learning. Cell 2016; 167:933-946.e20. [PMID: 27881303 DOI: 10.1016/j.cell.2016.09.055] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 07/21/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
To execute accurate movements, animals must continuously adapt their behavior to changes in their bodies and environments. Animals can learn changes in the relationship between their locomotor commands and the resulting distance moved, then adjust command strength to achieve a desired travel distance. It is largely unknown which circuits implement this form of motor learning, or how. Using whole-brain neuronal imaging and circuit manipulations in larval zebrafish, we discovered that the serotonergic dorsal raphe nucleus (DRN) mediates short-term locomotor learning. Serotonergic DRN neurons respond phasically to swim-induced visual motion, but little to motion that is not self-generated. During prolonged exposure to a given motosensory gain, persistent DRN activity emerges that stores the learned efficacy of motor commands and adapts future locomotor drive for tens of seconds. The DRN's ability to track the effectiveness of motor intent may constitute a computational building block for the broader functions of the serotonergic system. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Takashi Kawashima
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Maarten F Zwart
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Chao-Tsung Yang
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Brett D Mensh
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA.
| |
Collapse
|
37
|
Picton LD, Sillar KT. Mechanisms underlying the endogenous dopaminergic inhibition of spinal locomotor circuit function in Xenopus tadpoles. Sci Rep 2016; 6:35749. [PMID: 27760989 PMCID: PMC5071771 DOI: 10.1038/srep35749] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/30/2016] [Indexed: 01/08/2023] Open
Abstract
Dopamine plays important roles in the development and modulation of motor control circuits. Here we show that dopamine exerts potent effects on the central pattern generator circuit controlling locomotory swimming in post-embryonic Xenopus tadpoles. Dopamine (0.5–100 μM) reduced fictive swim bout occurrence and caused both spontaneous and evoked episodes to become shorter, slower and weaker. The D2-like receptor agonist quinpirole mimicked this repertoire of inhibitory effects on swimming, whilst the D4 receptor antagonist, L745,870, had the opposite effects. The dopamine reuptake inhibitor bupropion potently inhibited fictive swimming, demonstrating that dopamine constitutes an endogenous modulatory system. Both dopamine and quinpirole also inhibited swimming in spinalised preparations, suggesting spinally located dopamine receptors. Dopamine and quinpirole hyperpolarised identified rhythmically active spinal neurons, increased rheobase and reduced spike probability both during swimming and in response to current injection. The hyperpolarisation was TTX-resistant and was accompanied by decreased input resistance, suggesting that dopamine opens a K+ channel. The K+ channel blocker barium chloride (but not TEA, glybenclamide or tertiapin-Q) significantly occluded the hyperpolarisation. Overall, we show that endogenously released dopamine acts upon spinally located D2-like receptors, leading to a rapid inhibitory modulation of swimming via the opening of a K+ channel.
Collapse
Affiliation(s)
- Laurence D Picton
- School of Psychology and Neuroscience, University of St Andrews, St Andrews KY16 9JP, United Kingdom
| | - Keith T Sillar
- School of Psychology and Neuroscience, University of St Andrews, St Andrews KY16 9JP, United Kingdom
| |
Collapse
|
38
|
Chen YC, Semenova S, Rozov S, Sundvik M, Bonkowsky JL, Panula P. A Novel Developmental Role for Dopaminergic Signaling to Specify Hypothalamic Neurotransmitter Identity. J Biol Chem 2016; 291:21880-21892. [PMID: 27539857 DOI: 10.1074/jbc.m115.697466] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Indexed: 01/08/2023] Open
Abstract
Hypothalamic neurons expressing histamine and orexin/hypocretin (hcrt) are necessary for normal regulation of wakefulness. In Parkinson's disease, the loss of dopaminergic neurons is associated with elevated histamine levels and disrupted sleep/wake cycles, but the mechanism is not understood. To characterize the role of dopamine in the development of histamine neurons, we inhibited the translation of the two non-allelic forms of tyrosine hydroxylase (th1 and th2) in zebrafish larvae. We found that dopamine levels were reduced in both th1 and th2 knockdown, but the serotonin level and number of serotonin neurons remained unchanged. Further, we demonstrated that th2 knockdown increased histamine neuron number and histamine levels, whereas increased dopaminergic signaling using the dopamine precursor l-DOPA (l-3,4-dihydroxyphenylalanine) or dopamine receptor agonists reduced the number of histaminergic neurons. Increases in the number of histaminergic neurons were paralleled by matching increases in the numbers of hcrt neurons, supporting observations that histamine regulates hcrt neuron development. Finally, we show that histaminergic neurons surround th2-expressing neurons in the hypothalamus, and we suggest that dopamine regulates the terminal differentiation of histamine neurons via paracrine actions or direct synaptic neurotransmission. These results reveal a role for dopaminergic signaling in the regulation of neurotransmitter identity and a potential mechanism contributing to sleep disturbances in Parkinson's disease.
Collapse
Affiliation(s)
- Yu-Chia Chen
- From the Neuroscience Center and Department of Anatomy, University of Helsinki, 00290 Helsinki, Finland and
| | - Svetlana Semenova
- From the Neuroscience Center and Department of Anatomy, University of Helsinki, 00290 Helsinki, Finland and
| | - Stanislav Rozov
- From the Neuroscience Center and Department of Anatomy, University of Helsinki, 00290 Helsinki, Finland and
| | - Maria Sundvik
- From the Neuroscience Center and Department of Anatomy, University of Helsinki, 00290 Helsinki, Finland and
| | - Joshua L Bonkowsky
- the Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah 84113
| | - Pertti Panula
- From the Neuroscience Center and Department of Anatomy, University of Helsinki, 00290 Helsinki, Finland and
| |
Collapse
|
39
|
Robra L, Thirumalai V. The Intracellular Signaling Molecule Darpp-32 Is a Marker for Principal Neurons in the Cerebellum and Cerebellum-Like Circuits of Zebrafish. Front Neuroanat 2016; 10:81. [PMID: 27540357 PMCID: PMC4972821 DOI: 10.3389/fnana.2016.00081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/19/2016] [Indexed: 01/11/2023] Open
Abstract
The dopamine and cAMP regulated phosphoprotein of apparent molecular weight 32 kDa (Darpp-32) is an inhibitory subunit of protein phosphatase-1 (PP-1). Darpp-32 activity is regulated by multiple ligand-activated G-protein coupled receptors (GPCRs). This protein is coded for by the protein phosphatase-1 regulatory subunit 1b (ppp1r1b) gene. Here, we provide experimental evidence for the presence of multiple isoforms of ppp1r1b in zebrafish. We show that these isoforms are differentially expressed during development with the full-length isoform being maternally deposited. Next, with a custom polyclonal antibody generated against the full-length protein, we show that in the adult, Darpp-32 is strongly expressed in principal neurons of the cerebellum and cerebellum-like circuits. These include Purkinje neurons in the cerebellum, Type-I neurons in the optic tectum, and crest cells in the medial octavolateralis nucleus (MON). We confirmed the identity of these neurons through their colocalization with Parvalbumin 7 immunoreactivity. Darpp-32 is seen in the somata and dendrites of these neurons with faint staining in the axons. In all of these regions, Darpp-32-immunoreactive cells were in close proximity to tyrosine hydroxylase (TH) immunoreactive puncta indicating the presence of direct catecholaminergic input to these neurons. Darpp-32 immunoreactivity was seen in Purkinje neurons as early as 3 days post-fertilization (dpf) when Purkinje neurons are first specified. In sum, we show that Darpp-32, a signaling integrator, is a specific marker of principal neurons in the cerebellum and cerebellum-like circuits in zebrafish.
Collapse
Affiliation(s)
- Lena Robra
- National Centre for Biological Sciences Bangalore, India
| | | |
Collapse
|
40
|
Dukes AA, Bai Q, Van Laar VS, Zhou Y, Ilin V, David CN, Agim ZS, Bonkowsky JL, Cannon JR, Watkins SC, Croix CMS, Burton EA, Berman SB. Live imaging of mitochondrial dynamics in CNS dopaminergic neurons in vivo demonstrates early reversal of mitochondrial transport following MPP(+) exposure. Neurobiol Dis 2016; 95:238-49. [PMID: 27452482 DOI: 10.1016/j.nbd.2016.07.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/30/2016] [Accepted: 07/20/2016] [Indexed: 01/09/2023] Open
Abstract
Extensive convergent evidence collectively suggests that mitochondrial dysfunction is central to the pathogenesis of Parkinson's disease (PD). Recently, changes in the dynamic properties of mitochondria have been increasingly implicated as a key proximate mechanism underlying neurodegeneration. However, studies have been limited by the lack of a model in which mitochondria can be imaged directly and dynamically in dopaminergic neurons of the intact vertebrate CNS. We generated transgenic zebrafish in which mitochondria of dopaminergic neurons are labeled with a fluorescent reporter, and optimized methods allowing direct intravital imaging of CNS dopaminergic axons and measurement of mitochondrial transport in vivo. The proportion of mitochondria undergoing axonal transport in dopaminergic neurons decreased overall during development between 2days post-fertilization (dpf) and 5dpf, at which point the major period of growth and synaptogenesis of the relevant axonal projections is complete. Exposure to 0.5-1.0mM MPP(+) between 4 and 5dpf did not compromise zebrafish viability or cause detectable changes in the number or morphology of dopaminergic neurons, motor function or monoaminergic neurochemistry. However, 0.5mM MPP(+) caused a 300% increase in retrograde mitochondrial transport and a 30% decrease in anterograde transport. In contrast, exposure to higher concentrations of MPP(+) caused an overall reduction in mitochondrial transport. This is the first time mitochondrial transport has been observed directly in CNS dopaminergic neurons of a living vertebrate and quantified in a PD model in vivo. Our findings are compatible with a model in which damage at presynaptic dopaminergic terminals causes an early compensatory increase in retrograde transport of compromised mitochondria for degradation in the cell body. These data are important because manipulation of early pathogenic mechanisms might be a valid therapeutic approach to PD. The novel transgenic lines and methods we developed will be useful for future studies on mitochondrial dynamics in health and disease.
Collapse
Affiliation(s)
- April A Dukes
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Bai
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victor S Van Laar
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yangzhong Zhou
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Tsinghua University Medical School, Beijing, China
| | - Vladimir Ilin
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher N David
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; MSTP program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zeynep S Agim
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Simon C Watkins
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudette M St Croix
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA; Geriatric Research, Education and Clinical Center, Pittsburgh Veterans' Affairs Healthcare System, Pittsburgh, PA, USA.
| | - Sarah B Berman
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
41
|
Pantoja C, Hoagland A, Carroll EC, Karalis V, Conner A, Isacoff EY. Neuromodulatory Regulation of Behavioral Individuality in Zebrafish. Neuron 2016; 91:587-601. [PMID: 27397519 DOI: 10.1016/j.neuron.2016.06.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 05/05/2016] [Accepted: 06/08/2016] [Indexed: 11/30/2022]
Abstract
Inter-individual behavioral variation is thought to increase fitness and aid adaptation to environmental change, but the underlying mechanisms are poorly understood. We find that variation between individuals in neuromodulatory input contributes to individuality in short-term habituation of the zebrafish (Danio Rerio) acoustic startle response (ASR). ASR habituation varies greatly between individuals, but differences are stable over days and are heritable. Acoustic stimuli that activate ASR-command Mauthner cells also activate dorsal raphe nucleus (DRN) serotonergic neurons, which project to the vicinity of the Mauthner cells and their inputs. DRN neuron activity decreases during habituation in proportion to habituation and a genetic manipulation that reduces serotonin content in DRN neurons increases habituation, whereas serotonergic agonism or DRN activation with ChR2 reduces habituation. Finally, level of rundown of DRN activity co-segregates with extent of behavioral habituation across generations. Thus, variation between individuals in neuromodulatory input contributes to individuality in a core adaptive behavior. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Carlos Pantoja
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Adam Hoagland
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Elizabeth C Carroll
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Alden Conner
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
42
|
Ek F, Malo M, Åberg Andersson M, Wedding C, Kronborg J, Svensson P, Waters S, Petersson P, Olsson R. Behavioral Analysis of Dopaminergic Activation in Zebrafish and Rats Reveals Similar Phenotypes. ACS Chem Neurosci 2016; 7:633-46. [PMID: 26947759 DOI: 10.1021/acschemneuro.6b00014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Zebrafish is emerging as a complement to mammals in behavioral studies; however, there is a lack of comparative studies with rodents and humans to establish the zebrafish as a predictive translational model. Here we present a detailed phenotype evaluation of zebrafish larvae, measuring 300-3000 variables and analyzing them using multivariate analysis to identify the most important ones for further evaluations. The dopamine agonist apomorphine has previously been shown to have a complex U-shaped dose-response relationship in the variable distance traveled. In this study, we focused on breaking down distance traveled into more detailed behavioral phenotypes for both zebrafish and rats and identified in the multivariate analysis low and high dose phenotypes with characteristic behavioral features. Further analysis of single parameters also identified an increased activity at the lowest concentration indicative of a U-shaped dose-response. Apomorphine increased the distance of each swim movement (bout) at both high and low doses, but the underlying behavior of this increase is different; at high dose, both bout duration and frequency increased whereas bout max speed was higher at low dose. Larvae also displayed differences in place preference. The low dose phenotype spent more time in the center, indicative of an anxiolytic effect, while the high-dose phenotype had a wall preference. These dose-dependent effects corroborated findings in a parallel rat study and previous observations in humans. The translational value of pharmacological zebrafish studies was further evaluated by comparing the amino acid sequence of the dopamine receptors (D1-D4), between zebrafish, rats and humans. Humans and zebrafish share 100% of the amino acids in the binding site for D1 and D3 whereas D2 and D4 receptors share 85-95%. Molecular modeling of dopamine D2 and D4 receptors indicated that nonconserved amino acids have limited influence on important ligand-receptor interactions.
Collapse
Affiliation(s)
| | | | | | | | | | - Peder Svensson
- Integrative Research Laboratories Sweden AB, 413 46 Gothenburg, Sweden
| | - Susanna Waters
- Integrative Research Laboratories Sweden AB, 413 46 Gothenburg, Sweden
| | | | | |
Collapse
|
43
|
Dunn TW, Mu Y, Narayan S, Randlett O, Naumann EA, Yang CT, Schier AF, Freeman J, Engert F, Ahrens MB. Brain-wide mapping of neural activity controlling zebrafish exploratory locomotion. eLife 2016; 5:e12741. [PMID: 27003593 PMCID: PMC4841782 DOI: 10.7554/elife.12741] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/09/2016] [Indexed: 12/18/2022] Open
Abstract
In the absence of salient sensory cues to guide behavior, animals must still execute sequences of motor actions in order to forage and explore. How such successive motor actions are coordinated to form global locomotion trajectories is unknown. We mapped the structure of larval zebrafish swim trajectories in homogeneous environments and found that trajectories were characterized by alternating sequences of repeated turns to the left and to the right. Using whole-brain light-sheet imaging, we identified activity relating to the behavior in specific neural populations that we termed the anterior rhombencephalic turning region (ARTR). ARTR perturbations biased swim direction and reduced the dependence of turn direction on turn history, indicating that the ARTR is part of a network generating the temporal correlations in turn direction. We also find suggestive evidence for ARTR mutual inhibition and ARTR projections to premotor neurons. Finally, simulations suggest the observed turn sequences may underlie efficient exploration of local environments.
Collapse
Affiliation(s)
- Timothy W Dunn
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Program in Neuroscience, Department of Neurobiology, Harvard Medical School, Boston, United States.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Yu Mu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Sujatha Narayan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Owen Randlett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Eva A Naumann
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Chao-Tsung Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Program in Neuroscience, Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Jeremy Freeman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Program in Neuroscience, Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
44
|
Kishore S, McLean DL. Neuromodulation: letting sources of spinal dopamine speak for themselves. Curr Biol 2015; 25:R146-8. [PMID: 25689908 DOI: 10.1016/j.cub.2015.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A recent study of dopaminergic neurons in the brain of larval zebrafish has important implications for interpreting the natural actions of neuromodulators in the spinal cord.
Collapse
Affiliation(s)
- Sandeep Kishore
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - David L McLean
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
45
|
Anatomical Organization of Multiple Modulatory Inputs in a Rhythmic Motor System. PLoS One 2015; 10:e0142956. [PMID: 26566032 PMCID: PMC4643987 DOI: 10.1371/journal.pone.0142956] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/28/2015] [Indexed: 12/15/2022] Open
Abstract
In rhythmic motor systems, descending projection neuron inputs elicit distinct outputs from their target central pattern generator (CPG) circuits. Projection neuron activity is regulated by sensory inputs and inputs from other regions of the nervous system, relaying information about the current status of an organism. To gain insight into the organization of multiple inputs targeting a projection neuron, we used the identified neuron MCN1 in the stomatogastric nervous system of the crab, Cancer borealis. MCN1 originates in the commissural ganglion and projects to the stomatogastric ganglion (STG). MCN1 activity is differentially regulated by multiple inputs including neuroendocrine (POC) and proprioceptive (GPR) neurons, to elicit distinct outputs from CPG circuits in the STG. We asked whether these defined inputs are compact and spatially segregated or dispersed and overlapping relative to their target projection neuron. Immunocytochemical labeling, intracellular dye injection and three-dimensional (3D) confocal microscopy revealed overlap of MCN1 neurites and POC and GPR terminals. The POC neuron terminals form a defined neuroendocrine organ (anterior commissural organ: ACO) that utilizes peptidergic paracrine signaling to act on MCN1. The MCN1 arborization consistently coincided with the ACO structure, despite morphological variation between preparations. Contrary to a previous 2D study, our 3D analysis revealed that GPR axons did not terminate in a compact bundle, but arborized more extensively near MCN1, arguing against sparse connectivity of GPR onto MCN1. Consistent innervation patterns suggest that integration of the sensory GPR and peptidergic POC inputs occur through more distributed and more tightly constrained anatomical interactions with their common modulatory projection neuron target than anticipated.
Collapse
|
46
|
Montgomery JE, Wiggin TD, Rivera-Perez LM, Lillesaar C, Masino MA. Intraspinal serotonergic neurons consist of two, temporally distinct populations in developing zebrafish. Dev Neurobiol 2015; 76:673-87. [PMID: 26437856 DOI: 10.1002/dneu.22352] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 08/26/2015] [Accepted: 09/29/2015] [Indexed: 11/06/2022]
Abstract
Zebrafish intraspinal serotonergic neuron (ISN) morphology and distribution have been examined in detail at different ages; however, some aspects of the development of these cells remain unclear. Although antibodies to serotonin (5-HT) have detected ISNs in the ventral spinal cord of embryos, larvae, and adults, the only tryptophan hydroxylase (tph) transcript that has been described in the spinal cord is tph1a. Paradoxically, spinal tph1a is only expressed transiently in embryos, which brings the source of 5-HT in the ISNs of larvae and adults into question. Because the pet1 and tph2 promoters drive transgene expression in the spinal cord, we hypothesized that tph2 is expressed in spinal cords of zebrafish larvae. We confirmed this hypothesis through in situ hybridization. Next, we used 5-HT antibody labeling and transgenic markers of tph2-expressing neurons to identify a transient population of ISNs in embryos that was distinct from ISNs that appeared later in development. The existence of separate ISN populations may not have been recognized previously due to their shared location in the ventral spinal cord. Finally, we used transgenic markers and immunohistochemical labeling to identify the transient ISN population as GABAergic Kolmer-Agduhr double-prime (KA″) neurons. Altogether, this study revealed a novel developmental paradigm in which KA″ neurons are transiently serotonergic before the appearance of a stable population of tph2-expressing ISNs.
Collapse
Affiliation(s)
- Jacob E Montgomery
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Timothy D Wiggin
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Luis M Rivera-Perez
- Department of Biology, University of Puerto Rico in Ponce, Ponce, Puerto Rico
| | - Christina Lillesaar
- Department of Physiological Chemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - Mark A Masino
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
47
|
Ghahramani ZN, Timothy M, Kaur G, Gorbonosov M, Chernenko A, Forlano PM. Catecholaminergic Fiber Innervation of the Vocal Motor System Is Intrasexually Dimorphic in a Teleost with Alternative Reproductive Tactics. BRAIN, BEHAVIOR AND EVOLUTION 2015; 86:131-44. [PMID: 26355302 DOI: 10.1159/000438720] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/13/2015] [Indexed: 01/10/2023]
Abstract
Catecholamines, which include the neurotransmitters dopamine and noradrenaline, are known modulators of sensorimotor function, reproduction, and sexually motivated behaviors across vertebrates, including vocal-acoustic communication. Recently, we demonstrated robust catecholaminergic (CA) innervation throughout the vocal motor system in the plainfin midshipman fish Porichthys notatus, a seasonal breeding marine teleost that produces vocal signals for social communication. There are 2 distinct male reproductive morphs in this species: type I males establish nests and court females with a long-duration advertisement call, while type II males sneak spawn to steal fertilizations from type I males. Like females, type II males can only produce brief, agonistic, grunt type vocalizations. Here, we tested the hypothesis that intrasexual differences in the number of CA neurons and their fiber innervation patterns throughout the vocal motor pathway may provide neural substrates underlying divergence in reproductive behavior between morphs. We employed immunofluorescence (-ir) histochemistry to measure tyrosine hydroxylase (TH; a rate-limiting enzyme in catecholamine synthesis) neuron numbers in several forebrain and hindbrain nuclei as well as TH-ir fiber innervation throughout the vocal pathway in type I and type II males collected from nests during the summer reproductive season. After controlling for differences in body size, only one group of CA neurons displayed an unequivocal difference between male morphs: the extraventricular vagal-associated TH-ir neurons, located just lateral to the dimorphic vocal motor nucleus (VMN), were significantly greater in number in type II males. In addition, type II males exhibited greater TH-ir fiber density within the VMN and greater numbers of TH-ir varicosities with putative contacts on vocal motor neurons. This strong inverse relationship between the predominant vocal morphotype and the CA innervation of vocal motor neurons suggests that catecholamines may function to inhibit vocal output in midshipman. These findings support catecholamines as direct modulators of vocal behavior, and differential CA input appears reflective of social and reproductive behavioral divergence between male midshipman morphs.
Collapse
|
48
|
Filippi A, Mueller T, Driever W. vglut2 and gad expression reveal distinct patterns of dual GABAergic versus glutamatergic cotransmitter phenotypes of dopaminergic and noradrenergic neurons in the zebrafish brain. J Comp Neurol 2015; 522:2019-37. [PMID: 24374659 PMCID: PMC4288968 DOI: 10.1002/cne.23524] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 01/22/2023]
Abstract
Throughout the vertebrate lineage, dopaminergic neurons form important neuromodulatory systems that influence motor behavior, mood, cognition, and physiology. Studies in mammals have established that dopaminergic neurons often use γ-aminobutyric acid (GABA) or glutamatergic cotransmission during development and physiological function. Here, we analyze vglut2, gad1b and gad2 expression in combination with tyrosine hydroxylase immunoreactivity in 4-day-old larval and 30-day-old juvenile zebrafish brains to determine which dopaminergic and noradrenergic groups may use GABA or glutamate as a second transmitter. Our results show that most dopaminergic neurons also express GABAergic markers, including the dopaminergic groups of the olfactory bulb (homologous to mammalian A16) and the subpallium, the hypothalamic groups (A12, A14), the prethalamic zona incerta group (A13), the preoptic groups (A15), and the pretectal group. Thus, the majority of catecholaminergic neurons are gad1b/2-positive and coexpress GABA. A very few gad1/2-negative dopaminergic groups, however, express vglut2 instead and use glutamate as a second transmitter. These glutamatergic dual transmitter phenotypes are the Orthopedia transcription factor–dependent, A11-type dopaminergic neurons of the posterior tuberculum. All together, our results demonstrate that all catecholaminergic groups in zebrafish are either GABAergic or glutamatergic. Thus, cotransmission of dopamine and noradrenaline with either GABA or glutamate appears to be a regular feature of zebrafish catecholaminergic systems. We compare our results with those that have been described for mammalian systems, discuss the phenomenon of transmitter dualism in the context of developmental specification of GABAergic and glutamatergic regions in the brain, and put this phenomenon in an evolutionary perspective. J. Comp. Neurol. 522:2019–2037, 2014.
Collapse
Affiliation(s)
- Alida Filippi
- Developmental Biology, Institute of Biology I, Faculty of Biology, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | | | | |
Collapse
|
49
|
Herculano AM, Maximino C. Serotonergic modulation of zebrafish behavior: towards a paradox. Prog Neuropsychopharmacol Biol Psychiatry 2014; 55:50-66. [PMID: 24681196 DOI: 10.1016/j.pnpbp.2014.03.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 12/22/2022]
Abstract
Due to the fish-specific genome duplication event (~320-350 mya), some genes which code for serotonin proteins were duplicated in teleosts; this duplication event was preceded by a reorganization of the serotonergic system, with the appearance of the raphe nuclei (dependent on the isthmus organizer) and prosencephalic nuclei, including the paraventricular and pretectal complexes. With the appearance of amniotes, duplicated genes were lost, and the serotonergic system was reduced to a more complex raphe system. From a comparative point of view, then, the serotonergic system of zebrafish and that of mammals shows many important differences. However, many different behavioral functions of serotonin, as well as the effects of drugs which affect the serotonergic system, seem to be conserved among species. For example, in both zebrafish and rodents acute serotonin reuptake inhibitors (SSRIs) seem to increase anxiety-like behavior, while chronic SSRIs decrease it; drugs which act at the 5-HT1A receptor seem to decrease anxiety-like behavior in both zebrafish and rodents. In this article, we will expose this paradox, reviewing the chemical neuroanatomy of the zebrafish serotonergic system, followed by an analysis of the role of serotonin in zebrafish fear/anxiety, stress, aggression and the effects of psychedelic drugs.
Collapse
Affiliation(s)
- Anderson Manoel Herculano
- Neuroendocrinology Laboratory, Biological Sciences Institute, Federal University of Pará, Belém, PA, Brazil; "Frederico Graeff" Neurosciences and Behavior Laboratory, Department of Morphology and Physiological Sciences, Biological and Health Sciences Center, State University of Pará, Marabá, PA, Brazil
| | - Caio Maximino
- "Frederico Graeff" Neurosciences and Behavior Laboratory, Department of Morphology and Physiological Sciences, Biological and Health Sciences Center, State University of Pará, Marabá, PA, Brazil; International Zebrafish Neuroscience Research Consortium, United States.
| |
Collapse
|
50
|
Forlano PM, Kim SD, Krzyminska ZM, Sisneros JA. Catecholaminergic connectivity to the inner ear, central auditory, and vocal motor circuitry in the plainfin midshipman fish porichthys notatus. J Comp Neurol 2014; 522:2887-927. [PMID: 24715479 PMCID: PMC4107124 DOI: 10.1002/cne.23596] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 01/25/2023]
Abstract
Although the neuroanatomical distribution of catecholaminergic (CA) neurons has been well documented across all vertebrate classes, few studies have examined CA connectivity to physiologically and anatomically identified neural circuitry that controls behavior. The goal of this study was to characterize CA distribution in the brain and inner ear of the plainfin midshipman fish (Porichthys notatus) with particular emphasis on their relationship with anatomically labeled circuitry that both produces and encodes social acoustic signals in this species. Neurobiotin labeling of the main auditory end organ, the saccule, combined with tyrosine hydroxylase immunofluorescence (TH-ir) revealed a strong CA innervation of both the peripheral and central auditory system. Diencephalic TH-ir neurons in the periventricular posterior tuberculum, known to be dopaminergic, send ascending projections to the ventral telencephalon and prominent descending projections to vocal-acoustic integration sites, notably the hindbrain octavolateralis efferent nucleus, as well as onto the base of hair cells in the saccule via nerve VIII. Neurobiotin backfills of the vocal nerve in combination with TH-ir revealed CA terminals on all components of the vocal pattern generator, which appears to largely originate from local TH-ir neurons but may include input from diencephalic projections as well. This study provides strong neuroanatomical evidence that catecholamines are important modulators of both auditory and vocal circuitry and acoustic-driven social behavior in midshipman fish. This demonstration of TH-ir terminals in the main end organ of hearing in a nonmammalian vertebrate suggests a conserved and important anatomical and functional role for dopamine in normal audition.
Collapse
Affiliation(s)
- Paul M. Forlano
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY 11210
- Programs in Neuroscience, Ecology, Evolutionary Biology and Behavior, and Behavioral and Cognitive Neuroscience, The Graduate Center, City University of New York, Brooklyn, NY 11210
- Aquatic Research and Environmental Assessment Center, Brooklyn College, Brooklyn, NY
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - Spencer D. Kim
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY 11210
| | - Zuzanna M. Krzyminska
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY 11210
| | - Joseph A. Sisneros
- Departments of Psychology and Biology, University of Washington, Seattle, WA, 98195
- Virginia Merrill Bloedel Hearing Research Center, Seattle
- Marine Biological Laboratory, Woods Hole, MA 02543
| |
Collapse
|