1
|
Canzian J, Borba JV, Resmim CM, Mohammed KA, Pretzel CW, Adedara IA, Rosemberg DB. The dopamine transporter inhibition using GBR 12909 as a novel pharmacological tool to assess bipolar disorder-like neurobehavioral phenotypes in zebrafish. Behav Brain Res 2025; 477:115302. [PMID: 39442564 DOI: 10.1016/j.bbr.2024.115302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Dopamine (DA) is a neurotransmitter that plays an important role in brain physiology. Changes in DA-mediated signaling have been implicated with the pathophysiology of various neuropsychiatric conditions. Bipolar disorder (BD) is a mental disorder, characterized by alterning between manic/hypomanic and depressive mood. In experimental research, the pharmacological inhibition of DA reuptake using GBR 12909 serves as a tool to elicit BD-like phenotypes. Alternative model organisms, such as the zebrafish (Danio rerio), have been considered important systems for investigating the neurobehavioral changes involved in different neuropsychiatric conditions, including BD. Here, we discuss the use of GBR 12909 as a novel pharmacological strategy to mimic BD-like phenotypes in zebrafish models. We also emphasize the well-conserved DA-mediated signaling in zebrafish and the early expression of dopaminergic biomarkers in the brain, especially focusing on dopamine transporter (DAT), the main target of GBR 12909. Finally, we discuss potential advantages and limitations in the field, the perspectives of using GBR 12909 in BD research, and how distinct validation criteria (i.e., face, predictive, and construct validity) can be assessed in translational approaches using zebrafish-based models.
Collapse
Affiliation(s)
- Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.
| | - João V Borba
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Cássio M Resmim
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Khadija A Mohammed
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Camilla W Pretzel
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Isaac A Adedara
- Department of Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, Camobi, Santa Maria, RS 97105-900, Brazil
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
2
|
Hayashi S, Fujiuchi M, Oshiden M, Honda A, Kagawa N. Opioid receptor and dopaminergic gene expression increase in the brains of dominant medaka Oryzias latipes males after repeated fights. JOURNAL OF FISH BIOLOGY 2024. [PMID: 39462145 DOI: 10.1111/jfb.15980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The central opioid system and dopaminergic activity in mammals play key roles in mediating social reward, impulsivity, cognition, decision making, and motivation for learning and social interactions. Repeated positive fighting experiences enhance the gene expression levels of μ-type opioid receptor (Mor), tyrosine hydroxylase (Th), an enzyme involved in dopamine synthesis, and dopamine receptor type 2 (D2r) in the reward-related brain regions of aggressive mice. However, it remains unclear whether the opioid system and dopaminergic activity are associated with repeated winning in fish. In this study, we investigated changes in the expression levels of Mor, Th1, and D2r in different regions of the brain of adult medaka Oryzias latipes males after intermittent and continuous fight for 3 days. When a pair of males was provided a fighting opportunity for 20 min per day, we noted that within the 3-day observation period, aggressive winning males showed significantly higher expression levels of Mor in telencephalon and diencephalon, Th1 in diencephalon, and D2r in telencephalon than subordinate losing males. However, no such differences in gene expression level were observed between winning and losing males in the 3-day continuous fight. Further, no differences were detected in the total number of aggressive actions among the winners from each fighting test. However, the total number of "chase" actions, with a stronger aggressiveness index, was higher for the repeated winning male in the three-time intermittent fight than for the winner in the 3-day continuous fight. These findings suggest that repeated intermittent winning experiences with strong aggressiveness could be perceived as a reward by O. latipes males.
Collapse
Affiliation(s)
- Suzuna Hayashi
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| | - Miki Fujiuchi
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| | - Mei Oshiden
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| | - Akira Honda
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| | - Nao Kagawa
- Department of Life Science, Faculty of Science and Technology, Kindai University, Higashiosaka, Japan
| |
Collapse
|
3
|
Sagi D, Tibi M, Admati I, Lerer-Goldshtein T, Hochgerner H, Zeisel A, Appelbaum L. Single-Cell Profiling Uncovers Evolutionary Divergence of Hypocretin/Orexin Neuronal Subpopulations. J Neurosci 2024; 44:e0095242024. [PMID: 39122556 PMCID: PMC11376333 DOI: 10.1523/jneurosci.0095-24.2024] [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: 01/15/2024] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 08/12/2024] Open
Abstract
Brain nuclei are traditionally defined by their anatomy, activity, and expression of specific markers. The hypothalamus contains discrete neuronal populations that coordinate fundamental behavioral functions, including sleep and wakefulness, in all vertebrates. Particularly, the diverse roles of hypocretin/orexin (Hcrt)-releasing neurons suggest functional heterogeneity among Hcrt neurons. Using single-cell RNA sequencing (scRNA-seq) and high-resolution imaging of the adult male and female zebrafish hypothalamic periventricular zone, we identified 21 glutamatergic and 28 GABAergic cell types. Integration of zebrafish and mouse scRNA-seq revealed evolutionary conserved and divergent hypothalamic cell types. The expression of specific genes, including npvf, which encodes a sleep-regulating neuropeptide, was enriched in subsets of glutamatergic Hcrt neurons in both larval and adult zebrafish. The genetic profile, activity, and neurite processing of the neuronal subpopulation that coexpresses both Hcrt and Npvf (Hcrt+Npvf+) differ from other Hcrt neurons. These interspecies findings provide a unified annotation of hypothalamic cell types and suggest that the heterogeneity of Hcrt neurons enables multifunctionality, such as consolidation of both wake and sleep by the Hcrt- and Npvf-releasing neuronal subpopulation.
Collapse
Affiliation(s)
- Dana Sagi
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| | - Muhammad Tibi
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Inbal Admati
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Tali Lerer-Goldshtein
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| | - Hannah Hochgerner
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Amit Zeisel
- The Faculty of Biotechnology and Food Engineering, Technion 3200, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 590002, Israel
| |
Collapse
|
4
|
Kim MJ, Cho SH, Seo Y, Kim SD, Park HC, Kim BJ. Neuro-Restorative Effect of Nimodipine and Calcitriol in 1-Methyl 4-Phenyl 1,2,3,6 Tetrahydropyridine-Induced Zebrafish Parkinson's Disease Model. J Korean Neurosurg Soc 2024; 67:510-520. [PMID: 38130142 PMCID: PMC11375070 DOI: 10.3340/jkns.2023.0189] [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: 09/02/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
OBJECTIVE Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases, characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta. The treatment of PD aims to alleviate motor symptoms by replacing the reduced endogenous dopamine. Currently, there are no disease-modifying agents for the treatment of PD. Zebrafish (Danio rerio) have emerged as an effective tool for new drug discovery and screening in the age of translational research. The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is known to cause a similar loss of dopaminergic neurons in the human midbrain, with corresponding Parkinsonian symptoms. L-type calcium channels (LTCCs) have been implicated in the generation of mitochondrial oxidative stress, which underlies the pathogenesis of PD. Therefore, we investigated the neuro-restorative effect of LTCC inhibition in an MPTP-induced zebrafish PD model and suggested a possible drug candidate that might modify the progression of PD. METHODS All experiments were conducted using a line of transgenic zebrafish, Tg(dat:EGFP), in which green fluorescent protein (GFP) is expressed in dopaminergic neurons. The experimental groups were exposed to 500 μmol MPTP from 1 to 3 days post fertilization (dpf). The drug candidates : levodopa 1 mmol, nifedipine 10 μmol, nimodipine 3.5 μmol, diethylstilbestrol 0.3 μmol, luteolin 100 μmol, and calcitriol 0.25 μmol were exposed from 3 to 5 dpf. Locomotor activity was assessed by automated tracking and dopaminergic neurons were visualized in vivo by confocal microscopy. RESULTS Levodopa, nimodipine, diethylstilbestrol, and calcitriol had significant positive effects on the restoration of motor behavior, which was damaged by MPTP. Nimodipine and calcitriol have significant positive effects on the restoration of dopaminergic neurons, which were reduced by MPTP. Through locomotor analysis and dopaminergic neuron quantification, we identified the neuro-restorative effects of nimodipine and calcitriol in zebrafish MPTP-induced PD model. CONCLUSION The present study identified the neuro-restorative effects of nimodipine and calcitriol in an MPTP-induced zebrafish model of PD. They restored dopaminergic neurons which were damaged due to the effects of MPTP and normalized the locomotor activity. LTCCs have potential pathological roles in neurodevelopmental and neurodegenerative disorders. Zebrafish are highly amenable to high-throughput drug screening and might, therefore, be a useful tool to work towards the identification of diseasemodifying treatment for PD. Further studies including zebrafish genetic models to elucidate the mechanism of action of the diseasemodifying candidate by investigating Ca2+ influx and mitochondrial function in dopaminergic neurons, are needed to reveal the pathogenesis of PD and develop disease-modifying treatments for PD.
Collapse
Affiliation(s)
- Myung Ji Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| | - Su Hee Cho
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| | - Yongbo Seo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Sang-Dae Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Bum-Joon Kim
- Department of Neurosurgery, Ansan Hospital, Korea University Medical Center, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
5
|
Beauséjour PA, Veilleux JC, Condamine S, Zielinski BS, Dubuc R. Olfactory Projections to Locomotor Control Centers in the Sea Lamprey. Int J Mol Sci 2024; 25:9370. [PMID: 39273317 PMCID: PMC11395479 DOI: 10.3390/ijms25179370] [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: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Although olfaction is well known to guide animal behavior, the neural circuits underlying the motor responses elicited by olfactory inputs are poorly understood. In the sea lamprey, anatomical evidence shows that olfactory inputs project to the posterior tuberculum (PT), a structure containing dopaminergic (DA) neurons homologous to the mammalian ventral tegmental area and the substantia nigra pars compacta. Olfactory inputs travel directly from the medial olfactory bulb (medOB) or indirectly through the main olfactory bulb and the lateral pallium (LPal). Here, we characterized the transmission of olfactory inputs to the PT in the sea lamprey, Petromyzon marinus. Abundant projections from the medOB were observed close to DA neurons of the PT. Moreover, electrophysiological experiments revealed that PT neurons are activated by both the medOB and LPal, and calcium imaging indicated that the olfactory signal is then relayed to the mesencephalic locomotor region to initiate locomotion. In semi-intact preparations, stimulation of the medOB and LPal induced locomotion that was tightly associated with neural activity in the PT. Moreover, PT neurons were active throughout spontaneously occurring locomotor bouts. Altogether, our observations suggest that the medOB and LPal convey olfactory inputs to DA neurons of the PT, which in turn activate the brainstem motor command system to elicit locomotion.
Collapse
Affiliation(s)
| | - Jean-Christophe Veilleux
- Research Group in Adapted Physical Activity, Department of Exercise Sciences, Faculty of Sciences, University of Quebec in Montreal, Montreal, QC H2X 1Y4, Canada
| | - Steven Condamine
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Barbara S Zielinski
- Department of Integrative Biology, Faculty of Science, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Réjean Dubuc
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Group in Adapted Physical Activity, Department of Exercise Sciences, Faculty of Sciences, University of Quebec in Montreal, Montreal, QC H2X 1Y4, Canada
| |
Collapse
|
6
|
Hiraki-Kajiyama T, Miyasaka N, Ando R, Wakisaka N, Itoga H, Onami S, Yoshihara Y. An atlas and database of neuropeptide gene expression in the adult zebrafish forebrain. J Comp Neurol 2024; 532:e25619. [PMID: 38831653 DOI: 10.1002/cne.25619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 03/21/2024] [Accepted: 04/16/2024] [Indexed: 06/05/2024]
Abstract
Zebrafish is a useful model organism in neuroscience; however, its gene expression atlas in the adult brain is not well developed. In the present study, we examined the expression of 38 neuropeptides, comparing with GABAergic and glutamatergic neuron marker genes in the adult zebrafish brain by comprehensive in situ hybridization. The results are summarized as an expression atlas in 19 coronal planes of the forebrain. Furthermore, the scanned data of all brain sections were made publicly available in the Adult Zebrafish Brain Gene Expression Database (https://ssbd.riken.jp/azebex/). Based on these data, we performed detailed comparative neuroanatomical analyses of the hypothalamus and found that several regions previously described as one nucleus in the reference zebrafish brain atlas contain two or more subregions with significantly different neuropeptide/neurotransmitter expression profiles. Subsequently, we compared the expression data in zebrafish telencephalon and hypothalamus obtained in this study with those in mice, by performing a cluster analysis. As a result, several nuclei in zebrafish and mice were clustered in close vicinity. The present expression atlas, database, and anatomical findings will contribute to future neuroscience research using zebrafish.
Collapse
Affiliation(s)
- Towako Hiraki-Kajiyama
- Laboratory for Systems Molecular Ethology, RIKEN Center for Brain Science, Wako, Saitama, Japan
- Laboratory of Molecular Ethology, Graduate School of Life Science, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuhiko Miyasaka
- Laboratory for Systems Molecular Ethology, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Reiko Ando
- Support Unit for Bio-Material Analysis, Research Resources Division, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Noriko Wakisaka
- Laboratory for Systems Molecular Ethology, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Hiroya Itoga
- Laboratory for Developmental Dynamics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Shuichi Onami
- Laboratory for Developmental Dynamics, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Life Science Data Sharing Unit, RIKEN Information R&D and Strategy Headquarters, Kobe, Hyogo, Japan
| | - Yoshihiro Yoshihara
- Laboratory for Systems Molecular Ethology, RIKEN Center for Brain Science, Wako, Saitama, Japan
| |
Collapse
|
7
|
Hegarty BE, Gruenhagen GW, Johnson ZV, Baker CM, Streelman JT. Spatially resolved cell atlas of the teleost telencephalon and deep homology of the vertebrate forebrain. Commun Biol 2024; 7:612. [PMID: 38773256 PMCID: PMC11109250 DOI: 10.1038/s42003-024-06315-1] [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: 09/21/2023] [Accepted: 05/10/2024] [Indexed: 05/23/2024] Open
Abstract
The telencephalon has undergone remarkable diversification and expansion throughout vertebrate evolution, exhibiting striking variations in structural and functional complexity. Nevertheless, fundamental features are shared across vertebrate taxa, such as the presence of distinct regions including the pallium, subpallium, and olfactory structures. Teleost fishes have a uniquely "everted" telencephalon, which has confounded comparisons of their brain regions to other vertebrates. Here we combine spatial transcriptomics and single nucleus RNA-sequencing to generate a spatially-resolved transcriptional atlas of the Mchenga conophorus cichlid fish telencephalon. We then compare cell-types and anatomical regions in the cichlid telencephalon with those in amphibians, reptiles, birds, and mammals. We uncover striking transcriptional similarities between cell-types in the fish telencephalon and subpallial, hippocampal, and cortical cell-types in tetrapods, and find support for partial eversion of the teleost telencephalon. Ultimately, our work lends new insights into the organization and evolution of conserved cell-types and regions in the vertebrate forebrain.
Collapse
Affiliation(s)
- Brianna E Hegarty
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - George W Gruenhagen
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| | - Zachary V Johnson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, 30329, USA
| | - Cristina M Baker
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jeffrey T Streelman
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
8
|
Tanimoto Y, Kakinuma H, Aoki R, Shiraki T, Higashijima SI, Okamoto H. Transgenic tools targeting the basal ganglia reveal both evolutionary conservation and specialization of neural circuits in zebrafish. Cell Rep 2024; 43:113916. [PMID: 38484735 DOI: 10.1016/j.celrep.2024.113916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/18/2024] [Accepted: 02/17/2024] [Indexed: 04/02/2024] Open
Abstract
The cortico-basal ganglia circuit mediates decision making. Here, we generated transgenic tools for adult zebrafish targeting specific subpopulations of the components of this circuit and utilized them to identify evolutionary homologs of the mammalian direct- and indirect-pathway striatal neurons, which respectively project to the homologs of the internal and external segment of the globus pallidus (dorsal entopeduncular nucleus [dEN] and lateral nucleus of the ventral telencephalic area [Vl]) as in mammals. Unlike in mammals, the Vl mainly projects to the dEN directly, not by way of the subthalamic nucleus. Further single-cell RNA sequencing analysis reveals two pallidal output pathways: a major shortcut pathway directly connecting the dEN with the pallium and the evolutionarily conserved closed loop by way of the thalamus. Our resources and circuit map provide the common basis for the functional study of the basal ganglia in a small and optically tractable zebrafish brain for the comprehensive mechanistic understanding of the cortico-basal ganglia circuit.
Collapse
Affiliation(s)
- Yuki Tanimoto
- Laboratory for Neural Circuit Dynamics of Decision-making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Hisaya Kakinuma
- Laboratory for Neural Circuit Dynamics of Decision-making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Ryo Aoki
- Laboratory for Neural Circuit Dynamics of Decision-making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Toshiyuki Shiraki
- Research Resources Division, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Shin-Ichi Higashijima
- Exploratory Research Center on Life and Living Systems, Okazaki, Aichi 444-8787, Japan; National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | - Hitoshi Okamoto
- Laboratory for Neural Circuit Dynamics of Decision-making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; RIKEN CBS-Kao Collaboration Center, Saitama 351-0198, Japan.
| |
Collapse
|
9
|
Wullimann MF. Good heavens! Finally a landslide analysis of basal ganglia circuitry in teleosts. Cell Rep 2024; 43:113915. [PMID: 38484736 DOI: 10.1016/j.celrep.2024.113915] [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: 02/02/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 04/02/2024] Open
Abstract
Tanimoto et al.1 report essential information on teleostean basal ganglia circuitry. This analysis opens gateways into studying neurophysiology, neuropharmacology, and behavior in zebrafish, guided by this complex functional neural system common to all vertebrates.
Collapse
Affiliation(s)
- Mario F Wullimann
- Genes - Circuits - Behavior, Max-Planck-Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany; Department Biology II, Division of Neurobiology, Ludwig-Maximilians-University (LMU Munich), 82152 Martinsried, Germany.
| |
Collapse
|
10
|
Ilyin NP, Petersen EV, Kolesnikova TO, Demin KA, Khatsko SL, Apuhtin KV, Kalueff AV. Developing Peripheral Biochemical Biomarkers of Brain Disorders: Insights from Zebrafish Models. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:377-391. [PMID: 38622104 DOI: 10.1134/s0006297924020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/09/2024] [Accepted: 02/13/2024] [Indexed: 04/17/2024]
Abstract
High prevalence of human brain disorders necessitates development of the reliable peripheral biomarkers as diagnostic and disease-monitoring tools. In addition to clinical studies, animal models markedly advance studying of non-brain abnormalities associated with brain pathogenesis. The zebrafish (Danio rerio) is becoming increasingly popular as an animal model organism in translational neuroscience. These fish share some practical advantages over mammalian models together with high genetic homology and evolutionarily conserved biochemical and neurobehavioral phenotypes, thus enabling large-scale modeling of human brain diseases. Here, we review mounting evidence on peripheral biomarkers of brain disorders in zebrafish models, focusing on altered biochemistry (lipids, carbohydrates, proteins, and other non-signal molecules, as well as metabolic reactions and activity of enzymes). Collectively, these data strongly support the utility of zebrafish (from a systems biology standpoint) to study peripheral manifestations of brain disorders, as well as highlight potential applications of biochemical biomarkers in zebrafish models to biomarker-based drug discovery and development.
Collapse
Affiliation(s)
- Nikita P Ilyin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia.
| | - Elena V Petersen
- Moscow Institute of Physics and Technology, Moscow, 115184, Russia.
| | - Tatyana O Kolesnikova
- Neuroscience Program, Sirius University of Science and Technology, Sochi, 354340, Russia.
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia.
- Moscow Institute of Physics and Technology, Moscow, 115184, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of the Russian Federation, St. Petersburg, 197341, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of the Russian Federation, Pesochny, 197758, Russia
| | | | - Kirill V Apuhtin
- Laboratory of Biopsychiatry, Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, 630117, Russia.
- Neuroscience Division, Sirius University of Science and Technology, Sirius Federal Territory, 354340, Russia
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, 199034, Russia.
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of the Russian Federation, St. Petersburg, 197341, Russia
- Ural Federal University, Ekaterinburg, 620002, Russia
- Laboratory of Biopsychiatry, Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, 630117, Russia
| |
Collapse
|
11
|
Jin M, Shi R, Gao D, Wang B, Li N, Li X, Sik A, Liu K, Zhang X. ErbB2 pY -1248 as a predictive biomarker for Parkinson's disease based on research with RPPA technology and in vivo verification. CNS Neurosci Ther 2024; 30:e14407. [PMID: 37564024 PMCID: PMC10848095 DOI: 10.1111/cns.14407] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/12/2023] Open
Abstract
AIMS This study aims to reveal a promising biomarker for Parkinson's disease (PD) based on research with reverse phase protein array (RPPA) technology for the first time and in vivo verification, which gains time for early intervention in PD, thus increasing the effectiveness of treatment and reducing disease morbidity. METHODS AND RESULTS We employed RPPA technology which can assess both total and post-translationally modified proteins to identify biomarker candidates of PD in a cellular PD model. As a result, the phosphorylation (pY-1248) of the epidermal growth factor receptor (EGFR) ErbB2 is a promising biomarker candidate for PD. In addition, lapatinib, an ErbB2 tyrosine kinase inhibitor, was used to verify this PD biomarker candidate in vivo. We found that lapatinib-attenuated dopaminergic neuron loss and PD-like behavior in the zebrafish PD model. Accordingly, the expression of ErbB2pY-1248 significantly increased in the MPTP-induced mouse PD model. Our results suggest that ErbB2pY-1248 is a predictive biomarker for PD. CONCLUSIONS In this study, we found that ErbB2pY-1248 is a predictive biomarker of PD by using RPPA technology and in vivo verification. It offers a new perspective on PD diagnosing and treatment, which will be essential in identifying individuals at risk of PD. In addition, this study provides new ideas for digging into biomarkers of other neurodegenerative diseases.
Collapse
Affiliation(s)
- Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)Ji'nanChina
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong ProvinceJi'nanChina
| | - Ruidie Shi
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)Ji'nanChina
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong ProvinceJi'nanChina
- School of PsychologyNorth China University of Science and TechnologyTang'shanChina
| | - Daili Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)Ji'nanChina
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong ProvinceJi'nanChina
| | - Baokun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)Ji'nanChina
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong ProvinceJi'nanChina
| | - Ning Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)Ji'nanChina
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong ProvinceJi'nanChina
| | - Xia Li
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd.Ji'nanChina
| | - Attila Sik
- Institute of Transdisciplinary Discoveries, Medical SchoolUniversity of PecsPécsHungary
- Institute of Clinical Sciences, Medical SchoolUniversity of BirminghamBirminghamUK
- Institute of Physiology, Medical SchoolUniversity of PecsPécsHungary
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences)Ji'nanChina
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong ProvinceJi'nanChina
| | - Xiujun Zhang
- School of PsychologyNorth China University of Science and TechnologyTang'shanChina
| |
Collapse
|
12
|
Wiprich MT, da Rosa Vasques R, Gusso D, Rübensam G, Kist LW, Bogo MR, Bonan CD. Locomotor Behavior and Memory Dysfunction Induced by 3-Nitropropionic Acid in Adult Zebrafish: Modulation of Dopaminergic Signaling. Mol Neurobiol 2024; 61:609-621. [PMID: 37648841 DOI: 10.1007/s12035-023-03584-5] [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: 12/23/2022] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease characterized by neuropsychiatric disturbance, cognitive impairment, and locomotor dysfunction. In the early stage (chorea) of HD, expression of dopamine D2 receptors (D2R) is reduced, whereas dopamine (DA) levels are increased. Contrary, in the late stage (bradykinesia), DA levels and the expression of D2R and dopamine D1 receptors (D1R) are reduced. 3-Nitropropionic acid (3-NPA) is a toxin that may replicate HD behavioral phenotypes and biochemical aspects. This study assessed the neurotransmitter levels, dopamine receptor gene expression, and the effect of acute exposure to quinpirole (D2R agonist) and eticlopride (D2R antagonist) in an HD model induced by 3-NPA in adult zebrafish. Quinpirole and eticlopride were acutely applied by i.p. injection in adult zebrafish after chronic treatment of 3-NPA (60 mg/kg). 3-NPA treatment caused a reduction in DA, glutamate, and serotonin levels. Quinpirole reversed the bradykinesia and memory loss induced by 3-NPA. Together, these data showed that 3-NPA acts on the dopaminergic system and causes biochemical alterations similar to late-stage HD. These data reinforce the hypothesis that DA levels are linked with locomotor and memory deficits. Thus, these findings may suggest that the use of DA agonists could be a pharmacological strategy to improve the bradykinesia and memory deficits in the late-stage HD.
Collapse
Affiliation(s)
- Melissa Talita Wiprich
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Instituto Nacional de Ciência E Tecnologia Em Doenças Cerebrais, Excitotoxicidade E Neuroproteção, Porto Alegre, RS, Brazil
| | - Rafaela da Rosa Vasques
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Darlan Gusso
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Gabriel Rübensam
- Centro de Pesquisa Em Toxicologia E Farmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Luiza Wilges Kist
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Laboratório de Biologia Genômica E Molecular, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Mauricio Reis Bogo
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
- Laboratório de Biologia Genômica E Molecular, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Carla Denise Bonan
- Programa de Pós-Graduação Em Medicina E Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Laboratório de Neuroquímica E Psicofarmacologia, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
- Instituto Nacional de Ciência E Tecnologia Em Doenças Cerebrais, Excitotoxicidade E Neuroproteção, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Escola de Ciências da Saúde E da Vida, Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
13
|
Altbürger C, Rath M, Wehrle J, Driever W. The proneural factors Ascl1a and Ascl1b contribute to the terminal differentiation of dopaminergic GABAergic dual transmitter neurons in zebrafish. Dev Biol 2024; 505:58-74. [PMID: 37931393 DOI: 10.1016/j.ydbio.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/06/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023]
Abstract
The proneural factor Ascl1 is involved in several steps of neurogenesis, from neural progenitor maintenance to initiation of terminal differentiation and neuronal subtype specification. In neural progenitor cells, Ascl1 initiates the cell-cycle exit of progenitors, and contributes to their differentiation into mainly GABAergic neurons. Several catecholaminergic neuron groups in the forebrain of zebrafish use GABA as co-transmitter, but a potential role of the two paralogues Ascl1a and Ascl1b in their neurogenesis is not understood. Here, we show that ascl1a, ascl1b double mutant embryos develop a significantly reduced number of neurons in all GABAergic and catecholaminergic dual transmitter neuron anatomical clusters in the fore- and hindbrain, while glutamatergic catecholaminergic clusters develop normally. However, none of the affected catecholaminergic cell clusters are lost completely, suggesting an impairment in progenitor pools, or a requirement of Ascl1a/b for differentiation of a subset of neurons in each cluster. Early progenitors which are dlx2a+, fezf2 + or emx2 + are not reduced whereas late progenitors and differentiating neurons marked by the expression of dlx5a, isl1 and arxa are severely reduced in ascl1a, ascl1b double mutant embryos. This suggests that Ascl1a and Ascl1b play only a minor or no role in the maintenance of their progenitor pools, but rather contribute to the initiation of terminal differentiation of GABAergic catecholaminergic neurons.
Collapse
Affiliation(s)
- Christian Altbürger
- Department of Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University, Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany; CIBSS and BIOSS - Centres for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Meta Rath
- Department of Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University, Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany
| | - Johanna Wehrle
- Department of Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University, Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany; MeInBio Research Training Group, University of Freiburg, 79104, Freiburg, Germany
| | - Wolfgang Driever
- Department of Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University, Freiburg, Hauptstrasse 1, 79104, Freiburg, Germany; CIBSS and BIOSS - Centres for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany.
| |
Collapse
|
14
|
Roberta C, Vera S, Hans A H, Michael H H. Activation patterns of dopaminergic cell populations reflect different learning scenarios in a cichlid fish, Pseudotropheus zebra. J Chem Neuroanat 2023; 133:102342. [PMID: 37722435 DOI: 10.1016/j.jchemneu.2023.102342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/20/2023]
Abstract
Dopamine is present in all vertebrates and the functional roles of the subsystems are assumed to be similar. Whereas the effect of dopaminergic modulation is well investigated in different target systems, less is known about the factors that are causing the modulation of dopaminergic cells. Using the zebra mbuna, Pseudotropheus zebra, a cichlid fish from Lake Malawi as a model system, we investigated the activation of specific dopaminergic cell populations detected by double-labeling with TH and pS6 antibodies while the animals were solving different learning tasks. Specifically, we compared an intense avoidance learning situation, an instrumental learning task, and a non-learning isolated group and found strong activation of different dopaminergic cell populations. Preoptic-hypothalamic cell populations respond to the stress component in the avoidance task, and the forced movement/locomotion may be responsible for activation in the posterior tubercle. The instrumental learning task had little stress component, but the activation of the raphe superior in this group may be correlated with attention or arousal during the training sessions. At the same time, the weaker activation of the nucleus of the posterior commissure may be related to positive reward acting onto tectal circuits. Finally, we examined the co-activation patterns across all dopaminergic cell populations and recovered robust differences across experimental groups, largely driven by hypothalamic, posterior tubercle, and brain stem regions possibly encoding the valence and salience associated with stressful stimuli. Taken together, our results offer some insights into the different functions of the dopaminergic cell populations in the brain of a non-mammalian vertebrate in correlation with different behavioral conditions, extending our knowledge for a more comprehensive view of the mechanisms of dopaminergic modulation in vertebrates.
Collapse
Affiliation(s)
- Calvo Roberta
- Institute of Zoology, Rheinische Friedrich-Wilhelms-Universität Bonn, Poppelsdorfer Schloss, Meckenheimer Allee 169, 53115 Bonn, Germany.
| | - Schluessel Vera
- Institute of Zoology, Rheinische Friedrich-Wilhelms-Universität Bonn, Poppelsdorfer Schloss, Meckenheimer Allee 169, 53115 Bonn, Germany
| | - Hofmann Hans A
- Department of Integrative Biology, Institute for Neuroscience, University of Texas at Austin, 2415 Speedway, Austin, TX 78712, USA
| | - Hofmann Michael H
- Institute of Zoology, Rheinische Friedrich-Wilhelms-Universität Bonn, Poppelsdorfer Schloss, Meckenheimer Allee 169, 53115 Bonn, Germany
| |
Collapse
|
15
|
Toni M, Arena C, Cioni C, Tedeschi G. Temperature- and chemical-induced neurotoxicity in zebrafish. Front Physiol 2023; 14:1276941. [PMID: 37854466 PMCID: PMC10579595 DOI: 10.3389/fphys.2023.1276941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 09/22/2023] [Indexed: 10/20/2023] Open
Abstract
Throughout their lives, humans encounter a plethora of substances capable of inducing neurotoxic effects, including drugs, heavy metals and pesticides. Neurotoxicity manifests when exposure to these chemicals disrupts the normal functioning of the nervous system, and some neurotoxic agents have been linked to neurodegenerative pathologies such as Parkinson's and Alzheimer's disease. The growing concern surrounding the neurotoxic impacts of both naturally occurring and man-made toxic substances necessitates the identification of animal models for rapid testing across a wide spectrum of substances and concentrations, and the utilization of tools capable of detecting nervous system alterations spanning from the molecular level up to the behavioural one. Zebrafish (Danio rerio) is gaining prominence in the field of neuroscience due to its versatility. The possibility of analysing all developmental stages (embryo, larva and adult), applying the most common "omics" approaches (transcriptomics, proteomics, lipidomics, etc.) and conducting a wide range of behavioural tests makes zebrafish an excellent model for neurotoxicity studies. This review delves into the main experimental approaches adopted and the main markers analysed in neurotoxicity studies in zebrafish, showing that neurotoxic phenomena can be triggered not only by exposure to chemical substances but also by fluctuations in temperature. The findings presented here serve as a valuable resource for the study of neurotoxicity in zebrafish and define new scenarios in ecotoxicology suggesting that alterations in temperature can synergistically compound the neurotoxic effects of chemical substances, intensifying their detrimental impact on fish populations.
Collapse
Affiliation(s)
- Mattia Toni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Chiara Arena
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Carla Cioni
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science (DIVAS), Università Degli Studi di Milano, Milano, Italy
- CRC “Innovation for Well-Being and Environment” (I-WE), Università Degli Studi di Milano, Milano, Italy
| |
Collapse
|
16
|
Xu J, Casanave R, Chitre AS, Wang Q, Nguyen KM, Blake C, Wagle M, Cheng R, Polesskaya O, Palmer AA, Guo S. Causal Genetic Loci for a Motivated Behavior Spectrum Harbor Psychiatric Risk Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.06.556529. [PMID: 37732200 PMCID: PMC10508786 DOI: 10.1101/2023.09.06.556529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Behavioral diversity is critical for population fitness. Individual differences in risk-taking are observed across species, but underlying genetic mechanisms and conservation are largely unknown. We examined dark avoidance in larval zebrafish, a motivated behavior reflecting an approach-avoidance conflict. Brain-wide calcium imaging revealed significant neural activity differences between approach-inclined versus avoidance-inclined individuals. We used a population of ∼6,000 to perform the first genome-wide association study (GWAS) in zebrafish, which identified 34 genomic regions harboring many genes that are involved in synaptic transmission and human psychiatric diseases. We used CRISPR to study several causal genes: serotonin receptor-1b ( htr1b ), nitric oxide synthase-1 ( nos1 ), and stress-induced phosphoprotein-1 ( stip1 ). We further identified 52 conserved elements containing 66 GWAS significant variants. One encoded an exonic regulatory element that influenced tissue-specific nos1 expression. Together, these findings reveal new genetic loci and establish a powerful, scalable animal system to probe mechanisms underlying motivation, a critical dimension of psychiatric diseases.
Collapse
|
17
|
Altbürger C, Holzhauser J, Driever W. CRISPR/Cas9-based QF2 knock-in at the tyrosine hydroxylase ( th) locus reveals novel th-expressing neuron populations in the zebrafish mid- and hindbrain. Front Neuroanat 2023; 17:1196868. [PMID: 37603776 PMCID: PMC10433395 DOI: 10.3389/fnana.2023.1196868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/30/2023] [Indexed: 08/23/2023] Open
Abstract
Catecholaminergic neuron clusters are among the most conserved neuromodulatory systems in vertebrates, yet some clusters show significant evolutionary dynamics. Because of their disease relevance, special attention has been paid to mammalian midbrain dopaminergic systems, which have important functions in motor control, reward, motivation, and cognitive function. In contrast, midbrain dopaminergic neurons in teleosts were thought to be lost secondarily. Here, we generated a CRISPR/Cas9-based knock-in transgene at the th locus, which allows the expression of the Q-system transcription factor QF2 linked to the Tyrosine hydroxylase open reading frame by an E2A peptide. The QF2 knock-in allele still expresses Tyrosine hydroxylase in catecholaminergic neurons. Coexpression analysis of QF2 driven expression of QUAS fluorescent reporter transgenes and of th mRNA and Th protein revealed that essentially all reporter expressing cells also express Th/th. We also observed a small group of previously unidentified cells expressing the reporter gene in the midbrain and a larger group close to the midbrain-hindbrain boundary. However, we detected no expression of the catecholaminergic markers ddc, slc6a3, or dbh in these neurons, suggesting that they are not actively transmitting catecholamines. The identified neurons in the midbrain are located in a GABAergic territory. A coexpression analysis with anatomical markers revealed that Th-expressing neurons in the midbrain are located in the tegmentum and those close to the midbrain-hindbrain boundary are located in the hindbrain. Our data suggest that zebrafish may still have some evolutionary remnants of midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- Christian Altbürger
- Developmental Biology, Faculty of Biology, Institute of Biology I, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS and BIOSS - Centres for Biological Signalling Studies, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Jens Holzhauser
- Developmental Biology, Faculty of Biology, Institute of Biology I, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Faculty of Biology, Institute of Biology I, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS and BIOSS - Centres for Biological Signalling Studies, Albert Ludwigs University Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Hegarty BE, Gruenhagen GW, Johnson ZV, Baker CM, Streelman JT. Spatially resolved cell atlas of the teleost telencephalon and deep homology of the vertebrate forebrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549873. [PMID: 37503039 PMCID: PMC10370212 DOI: 10.1101/2023.07.20.549873] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The telencephalon has undergone remarkable diversification and expansion throughout vertebrate evolution, exhibiting striking differences in structural and functional complexity. Nevertheless, fundamental features are shared across vertebrate taxa, such as the presence of distinct regions including the pallium, subpallium, and olfactory structures. Teleost fishes have a uniquely 'everted' telencephalon, which has made it challenging to compare brain regions in fish to those in other vertebrates. Here we combine spatial transcriptomics and single-nucleus RNA-sequencing to generate a spatially-resolved transcriptional atlas of the cichlid fish telencephalon. We then compare cell-types and anatomical regions in the cichlid telencephalon with those in amphibians, reptiles, birds, and mammals. We uncover striking transcriptional similarities between cell populations in the fish telencephalon and subpallial, hippocampal, and cortical cell populations in tetrapods. Ultimately, our work lends new insights into the organization and evolution of conserved cell-types and regions in the vertebrate forebrain.
Collapse
Affiliation(s)
- Brianna E Hegarty
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - George W Gruenhagen
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Zachary V Johnson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30329
| | - Cristina M Baker
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jeffrey T Streelman
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332
- Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
19
|
Natsaridis E, Perdikaris P, Fokos S, Dermon CR. Neuronal and Astroglial Localization of Glucocorticoid Receptor GRα in Adult Zebrafish Brain ( Danio rerio). Brain Sci 2023; 13:861. [PMID: 37371341 DOI: 10.3390/brainsci13060861] [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: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Glucocorticoid receptor α (GRα), a ligand-regulated transcription factor, mainly activated by cortisol in humans and fish, mediates neural allostatic and homeostatic functions induced by different types of acute and chronic stress, and systemic inflammation. Zebrafish GRα is suggested to have multiple transcriptional effects essential for normal development and survival, similarly to mammals. While sequence alignments of human, monkey, rat, and mouse GRs have shown many GRα isoforms, we questioned the protein expression profile of GRα in the adult zebrafish (Danio rerio) brain using an alternative model for stress-related neuropsychiatric research, by means of Western blot, immunohistochemistry and double immunofluorescence. Our results identified four main GRα-like immunoreactive bands (95 kDa, 60 kDa, 45 kDa and 35 kDa), with the 95 kDa protein showing highest expression in forebrain compared to midbrain and hindbrain. GRα showed a wide distribution throughout the antero-posterior zebrafish brain axis, with the most prominent labeling within the telencephalon, preoptic, hypothalamus, midbrain, brain stem, central grey, locus coeruleus and cerebellum. Double immunofluorescence revealed that GRα is coexpressed in TH+, β2-AR+ and vGLUT+ neurons, suggesting the potential of GRα influences on adrenergic and glutamatergic transmission. Moreover, GRα was co-localized in midline astroglial cells (GFAP+) within the telencephalon, hypothalamus and hindbrain. Interestingly, GRα expression was evident in the brain regions involved in adaptive stress responses, social behavior, and sensory and motor integration, supporting the evolutionarily conserved features of glucocorticoid receptors in the zebrafish brain.
Collapse
Affiliation(s)
- Evangelos Natsaridis
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Panagiotis Perdikaris
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Stefanos Fokos
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| | - Catherine R Dermon
- Laboratory of Human and Animal Physiology, Department of Biology, University of Patras, Rion, 26504 Patras, Greece
| |
Collapse
|
20
|
Briñez-Gallego P, da Costa Silva DG, Cordeiro MF, Horn AP, Hort MA. Experimental models of chemically induced Parkinson's disease in zebrafish at the embryonic larval stage: a systematic review. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:201-237. [PMID: 36859813 DOI: 10.1080/10937404.2023.2182390] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra that results in a decrease in dopamine levels, resulting in motor-type disturbances. Different vertebrate models, such as rodents and fish, have been used to study PD. In recent decades, Danio rerio (zebrafish) has emerged as a potential model for the investigation of neurodegenerative diseases due to its homology to the nervous system of humans. In this context, this systematic review aimed to identify publications that reported the utilization of neurotoxins as an experimental model of parkinsonism in zebrafish embryos and larvae. Ultimately, 56 articles were identified by searching three databases (PubMed, Web of Science, and Google Scholar). Seventeen studies using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 4 1-methyl-4-phenylpyridinium (MPP+), 24 6-hydroxydopamine (6-OHDA), 6 paraquat/diquat, 2 rotenone, and 6 articles using other types of unusual neurotoxins to induce PD were selected. Neurobehavioral function, such as motor activity, dopaminergic neuron markers, oxidative stress biomarkers, and other relevant parameters in the zebrafish embryo-larval model were examined. In summary, this review provides information to help researchers determine which chemical model is suitable to study experimental parkinsonism, according to the effects induced by neurotoxins in zebrafish embryos and larvae.
Collapse
Affiliation(s)
- Paola Briñez-Gallego
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, RS, Brasil
| | - Dennis Guilherme da Costa Silva
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, RS, Brasil
| | - Marcos Freitas Cordeiro
- Programa de Pós-graduação em Biociências e Saúde, Universidade do Oeste de Santa Catarina - UNOESC, Joaçaba, SC, Brasil
| | - Ana Paula Horn
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, RS, Brasil
| | - Mariana Appel Hort
- Programa de Pós-graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, RS, Brasil
| |
Collapse
|
21
|
Yamaguchi Y, Takagi W, Kaiya H, Konno N, Yoshida MA, Kuraku S, Hyodo S. Phylogenetic and functional properties of hagfish neurohypophysial hormone receptors distinct from their jawed vertebrate counterparts. Gen Comp Endocrinol 2023; 336:114257. [PMID: 36868365 DOI: 10.1016/j.ygcen.2023.114257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Vertebrate neurohypophysial hormones, i.e., vasopressin- and oxytocin-family peptides, exert versatile physiological actions via distinct G protein-coupled receptors. The neurohypophysial hormone receptor (NHR) family was classically categorized into four subtypes (V1aR, V1bR, V2R and OTR), while recent studies have identified seven subtypes (V1aR, V1bR, V2aR, V2bR, V2cR, V2dR and OTR; V2aR corresponds to the conventional V2R). The vertebrate NHR family were diversified via multiple gene duplication events at different scales. Despite intensive research effort in non-osteichthyes vertebrates such as cartilaginous fish and lamprey, the molecular phylogeny of the NHR family has not been fully understood. In the present study, we focused on the inshore hagfish (Eptatretus burgeri), another group of cyclostomes, and Arctic lamprey (Lethenteron camtschaticum) for comparison. Two putative NHR homologs, which were previously identified only in silico, were cloned from the hagfish and designated as ebV1R and ebV2R. In vitro, ebV1R, as well as two out of five Arctic lamprey NHRs, increased intracellular Ca2+ in response to exogenous neurohypophysial hormones. None of the examined cyclostome NHRs altered intracellular cAMP levels. Transcripts of ebV1R were detected in multiple tissues including the brain and gill, with intense hybridization signals in the hypothalamus and adenohypophysis, while ebV2R was predominantly expressed in the systemic heart. Similarly, Arctic lamprey NHRs showed distinct expression patterns, underscoring the multifunctionality of VT in the cyclostomes as in the gnathostomes. These results and exhaustive gene synteny comparisons provide new insights into the molecular and functional evolution of the neurohypophysial hormone system in vertebrates.
Collapse
Affiliation(s)
- Yoko Yamaguchi
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan.
| | - Wataru Takagi
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, Osaka 564-8565, Japan; Grandsoul Research Institute for Immunology, Inc., Matsui 8-1 Utano, Uda, Nara 633-2221, Japan
| | - Norifumi Konno
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Masa-Aki Yoshida
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan; Marine Biological Science Section, Education and Research Center for Biological Resources, Faculty of Life and Environmental Science, Shimane University, 194 Kamo, Okinoshima, Oki, Shimane 685-0024, Japan
| | - Shigehiro Kuraku
- Molecular Life History Laboratory, Department of Genomics and Evolutionary Biology, National Institution of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, Sokendai (Graduate University for Advanced Studies), 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojimaminami-machi, Chuo-ku, Kobe 650-0047, Japan
| | - Susumu Hyodo
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8564, Japan
| |
Collapse
|
22
|
Allen A, Heisler E, Kittelberger JM. Dopamine injections to the midbrain periaqueductal gray inhibit vocal-motor production in a teleost fish. Physiol Behav 2023; 263:114131. [PMID: 36796532 DOI: 10.1016/j.physbeh.2023.114131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
Across vertebrates, the midbrain periaqueductal gray (PAG) plays a critical role in social and vocal behavior. Dopaminergic neurotransmission also modulates these behaviors, and dopaminergic innervation of the PAG has been well documented. Nonetheless, the potential role of dopamine in shaping vocal production at the level of the PAG is not well understood. Here, we tested the hypothesis that dopamine modulates vocal production in the PAG, using a well-characterized vertebrate model system for the study of vocal communication, the plainfin midshipman fish, Porichthys notatus. We found that focal dopamine injections to the midshipman PAG rapidly and reversibly inhibited vocal production triggered by stimulation of known vocal-motor structures in the preoptic area / anterior hypothalamus. While dopamine inhibited vocal-motor output, it did not alter behaviorally-relevant parameters of this output, such as vocalization duration and frequency. Dopamine-induced inhibition of vocal production was prevented by the combined blockade of D1- and D2-like receptors but was unaffected by isolated blockade of either D1-receptors or D2-receptors. Our results suggest dopamine neuromodulation in the midshipman PAG may inhibit natural vocal behavior, in courtship and/or agonistic social contexts.
Collapse
Affiliation(s)
- Alexander Allen
- Department of Biology, Gettysburg College, Gettysburg, PA 17325, United States
| | - Elizabeth Heisler
- Department of Biology, Gettysburg College, Gettysburg, PA 17325, United States
| | | |
Collapse
|
23
|
Esancy K, Conceicao LL, Curtright A, Tran T, Condon L, Lecamp B, Dhaka A. A novel small molecule, AS1, reverses the negative hedonic valence of noxious stimuli. BMC Biol 2023; 21:69. [PMID: 37013580 PMCID: PMC10071644 DOI: 10.1186/s12915-023-01573-7] [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: 10/03/2022] [Accepted: 03/17/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Pain is the primary reason people seek medical care, with chronic pain affecting ~ 20% of people in the USA. However, many existing analgesics are ineffective in treating chronic pain, while others (e.g., opioids) have undesirable side effects. Here, we describe the screening of a small molecule library using a thermal place aversion assay in larval zebrafish to identify compounds that alter aversion to noxious thermal stimuli and could thus serve as potential analgesics. RESULTS From our behavioral screen, we discovered a small molecule, Analgesic Screen 1 (AS1), which surprisingly elicited attraction to noxious painful heat. When we further explored the effects of this compound using other behavioral place preference assays, we found that AS1 was similarly able to reverse the negative hedonic valence of other painful (chemical) and non-painful (dark) aversive stimuli without being inherently rewarding. Interestingly, targeting molecular pathways canonically associated with analgesia did not replicate the effects of AS1. A neuronal imaging assay revealed that clusters of dopaminergic neurons, as well as forebrain regions located in the teleost equivalent of the basal ganglia, were highly upregulated in the specific context of AS1 and aversive heat. Through a combination of behavioral assays and pharmacological manipulation of dopamine circuitry, we determined that AS1 acts via D1 dopamine receptor pathways to elicit this attraction to noxious stimuli. CONCLUSIONS Together, our results suggest that AS1 relieves an aversion-imposed "brake" on dopamine release, and that this unique mechanism may provide valuable insight into the development of new valence-targeting analgesic drugs, as well as medications for other valence-related neurological conditions, such as anxiety and post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Kali Esancy
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Lais L Conceicao
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Andrew Curtright
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Thanh Tran
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Logan Condon
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Bryce Lecamp
- Department of Biological Structure, University of Washington, Seattle, USA
| | - Ajay Dhaka
- Department of Biological Structure, University of Washington, Seattle, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, USA.
| |
Collapse
|
24
|
Weinschutz Mendes H, Neelakantan U, Liu Y, Fitzpatrick SE, Chen T, Wu W, Pruitt A, Jin DS, Jamadagni P, Carlson M, Lacadie CM, Enriquez KD, Li N, Zhao D, Ijaz S, Sakai C, Szi C, Rooney B, Ghosh M, Nwabudike I, Gorodezky A, Chowdhury S, Zaheer M, McLaughlin S, Fernandez JM, Wu J, Eilbott JA, Vander Wyk B, Rihel J, Papademetris X, Wang Z, Hoffman EJ. High-throughput functional analysis of autism genes in zebrafish identifies convergence in dopaminergic and neuroimmune pathways. Cell Rep 2023; 42:112243. [PMID: 36933215 PMCID: PMC10277173 DOI: 10.1016/j.celrep.2023.112243] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/15/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
Advancing from gene discovery in autism spectrum disorders (ASDs) to the identification of biologically relevant mechanisms remains a central challenge. Here, we perform parallel in vivo functional analysis of 10 ASD genes at the behavioral, structural, and circuit levels in zebrafish mutants, revealing both unique and overlapping effects of gene loss of function. Whole-brain mapping identifies the forebrain and cerebellum as the most significant contributors to brain size differences, while regions involved in sensory-motor control, particularly dopaminergic regions, are associated with altered baseline brain activity. Finally, we show a global increase in microglia resulting from ASD gene loss of function in select mutants, implicating neuroimmune dysfunction as a key pathway relevant to ASD biology.
Collapse
Affiliation(s)
| | - Uma Neelakantan
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Sarah E Fitzpatrick
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; MD-PhD Program, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianying Chen
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Weimiao Wu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - April Pruitt
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA
| | - David S Jin
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Marina Carlson
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA
| | - Cheryl M Lacadie
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Ningshan Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA; SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dejian Zhao
- Department of Genetics, Yale Center for Genome Analysis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sundas Ijaz
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Catalina Sakai
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christina Szi
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Brendan Rooney
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Marcus Ghosh
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ijeoma Nwabudike
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; MD-PhD Program, Yale School of Medicine, New Haven, CT 06510, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrea Gorodezky
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sumedha Chowdhury
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Meeraal Zaheer
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sarah McLaughlin
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Jia Wu
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jeffrey A Eilbott
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Brent Vander Wyk
- Department of Internal Medicine, Section of Geriatrics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Xenophon Papademetris
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06510, USA
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT 06510, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
25
|
Nabinger DD, Altenhofen S, Buatois A, Facciol A, Peixoto JV, da Silva JMK, Chatterjee D, Rübensam G, Gerlai R, Bonan CD. Acute administration of a dopamine D2/D3 receptor agonist alters behavioral and neural parameters in adult zebrafish. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110753. [PMID: 36934998 DOI: 10.1016/j.pnpbp.2023.110753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/30/2023] [Accepted: 03/15/2023] [Indexed: 03/21/2023]
Abstract
The dopaminergic neurotransmitter system is implicated in several brain functions and behavioral processes. Alterations in it are associated with the pathogenesis of several human neurological disorders. Pharmacological agents that interact with the dopaminergic system allow the investigation of dopamine-mediated cellular and molecular responses and may elucidate the biological bases of such disorders. Zebrafish, a translationally relevant biomedical research organism, has been successfully employed in prior psychopharmacology studies. Here, we evaluated the effects of quinpirole (dopamine D2/D3 receptor agonist) in adult zebrafish on behavioral parameters, brain-derived neurotrophic factor (BDNF) and neurotransmitter levels. Zebrafish received intraperitoneal injections of 0.5, 1.0, or 2.0 mg/kg quinpirole or saline (control group) twice with an inter-injection interval of 48 h. All tests were performed 24 h after the second injection. After this acute quinpirole administration, zebrafish exhibited decreased locomotor activity, increased anxiety-like behaviors and memory impairment. However, quinpirole did not affect social and aggressive behavior. Quinpirole-treated fish exhibited stereotypic swimming, characterized by repetitive behavior followed by immobile episodes. Moreover, quinpirole treatment also decreased the number of BDNF-immunoreactive cells in the zebrafish brain. Analysis of neurotransmitter levels demonstrated a significant increase in glutamate and a decrease in serotonin, while no alterations were observed in dopamine. These findings demonstrate that dopaminergic signaling altered by quinpirole administration results in significant behavioral and neuroplastic changes in the central nervous system of zebrafish. Thus, we conclude that the use of quinpirole administration in adult zebrafish may be an appropriate tool for the analysis of mechanisms underlying neurological disorders related to the dopaminergic system.
Collapse
Affiliation(s)
- Débora Dreher Nabinger
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Stefani Altenhofen
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexis Buatois
- Department of Psychology, University of Toronto Mississauga, ON, Canada
| | - Amanda Facciol
- Department of Psychology, University of Toronto Mississauga, ON, Canada
| | - Julia Vasconcellos Peixoto
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Julia Maria Kuhl da Silva
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Gabriel Rübensam
- Centro de Pesquisa em Toxicologia e Farmacologia (INTOX), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Robert Gerlai
- Department of Psychology, University of Toronto Mississauga, ON, Canada
| | - Carla Denise Bonan
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Cerebrais, Excitotoxicidade e Neuroproteção, Porto Alegre, RS, Brazil.
| |
Collapse
|
26
|
Peña-Díaz S, García-Pardo J, Ventura S. Development of Small Molecules Targeting α-Synuclein Aggregation: A Promising Strategy to Treat Parkinson's Disease. Pharmaceutics 2023; 15:839. [PMID: 36986700 PMCID: PMC10059018 DOI: 10.3390/pharmaceutics15030839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Parkinson's disease, the second most common neurodegenerative disorder worldwide, is characterized by the accumulation of protein deposits in the dopaminergic neurons. These deposits are primarily composed of aggregated forms of α-Synuclein (α-Syn). Despite the extensive research on this disease, only symptomatic treatments are currently available. However, in recent years, several compounds, mainly of an aromatic character, targeting α-Syn self-assembly and amyloid formation have been identified. These compounds, discovered by different approaches, are chemically diverse and exhibit a plethora of mechanisms of action. This work aims to provide a historical overview of the physiopathology and molecular aspects associated with Parkinson's disease and the current trends in small compound development to target α-Syn aggregation. Although these molecules are still under development, they constitute an important step toward discovering effective anti-aggregational therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Samuel Peña-Díaz
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Javier García-Pardo
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
27
|
Burgess HA, Burton EA. A Critical Review of Zebrafish Neurological Disease Models-1. The Premise: Neuroanatomical, Cellular and Genetic Homology and Experimental Tractability. OXFORD OPEN NEUROSCIENCE 2023; 2:kvac018. [PMID: 37649777 PMCID: PMC10464506 DOI: 10.1093/oons/kvac018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/13/2022] [Indexed: 09/01/2023]
Abstract
The last decade has seen a dramatic rise in the number of genes linked to neurological disorders, necessitating new models to explore underlying mechanisms and to test potential therapies. Over a similar period, many laboratories adopted zebrafish as a tractable model for studying brain development, defining neural circuits and performing chemical screens. Here we discuss strengths and limitations of using the zebrafish system to model neurological disorders. The underlying premise for many disease models is the high degree of homology between human and zebrafish genes, coupled with the conserved vertebrate Bauplan and repertoire of neurochemical signaling molecules. Yet, we caution that important evolutionary divergences often limit the extent to which human symptoms can be modeled meaningfully in zebrafish. We outline advances in genetic technologies that allow human mutations to be reproduced faithfully in zebrafish. Together with methods that visualize the development and function of neuronal pathways at the single cell level, there is now an unprecedented opportunity to understand how disease-associated genetic changes disrupt neural circuits, a level of analysis that is ideally suited to uncovering pathogenic changes in human brain disorders.
Collapse
Affiliation(s)
- Harold A Burgess
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20892, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, 15260, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA,15260, USA
- Geriatric Research, Education, and Clinical Center, Pittsburgh VA Healthcare System, Pittsburgh, PA, 15240, USA
| |
Collapse
|
28
|
Afferent and efferent connections of the nucleus posterior tuberis in the firemouth cichlid, Thorichthys meeki. Neurosci Res 2023; 186:10-20. [PMID: 36007624 DOI: 10.1016/j.neures.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/14/2022] [Accepted: 08/18/2022] [Indexed: 01/04/2023]
Abstract
The nucleus posterior tuberis (NPT) in teleost fishes, also called posterior tuberal nucleus, is situated in the posterior tuberculum of the diencephalon. It is fused across the midline and densely packed with small cells, but little is known about its connections. In this study, the afferent and efferent connections of the NPT were examined by means of tracer applications of the carbocyanine dye DiI in the firemouth cichlid, Thorichthys meeki. Retrogradely labeled cell bodies were found in the corpus mamillare and nucleus periventricularis of the inferior lobe; and anterogradely labeled terminal fibers were detected in the medial zone of the dorsal telencephalon, medial part of the nucleus lateralis tuberis, dorsal posterior thalamic nucleus, torus lateralis, medial part of the nucleus diffusus of the inferior lobe, and tectum opticum. All these connections show an ipsilateral tendency. The NPT is apparently a significant relay nucleus in the diencephalon of T. meeki, and possibly involved in a variety of feedback circuits. It seems also to be part of a tecto-hypothalamo-telencephalic pathway in cichlids.
Collapse
|
29
|
Hettiarachchi P, Niyangoda SS, Jarosova R, Johnson MA. Dopamine Release Impairments Accompany Locomotor and Cognitive Deficiencies in Rotenone-Treated Parkinson's Disease Model Zebrafish. Chem Res Toxicol 2022; 35:1974-1982. [PMID: 36178476 PMCID: PMC10127151 DOI: 10.1021/acs.chemrestox.2c00150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this work, we carried out neurochemical and behavioral analysis of zebrafish (Danio rerio) treated with rotenone, an agent used to chemically induce a syndrome resembling Parkinson's disease (PD). Dopamine release, measured with fast-scan cyclic voltammetry (FSCV) at carbon-fiber electrodes in acutely harvested whole brains, was about 30% of that found in controls. Uptake, represented by the first order rate constant (k) and the half-life (t1/2) determined by nonlinear regression modeling of the stimulated release plots, was also diminished. Behavioral analysis revealed that rotenone treatment increased the time required for zebrafish to reach a reward within a maze by more than 50% and caused fish to select the wrong pathway, suggesting that latent learning was impaired. Additionally, zebrafish treated with rotenone suffered from diminished locomotor activity, swimming shorter distances with lower mean velocity and acceleration. Thus, the neurochemical and behavioral approaches, as applied, were able to resolve rotenone-induced differences in key parameters. This approach may be effective for screening therapies in this and other models of neurodegeneration.
Collapse
Affiliation(s)
- Piyanka Hettiarachchi
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Sayuri S. Niyangoda
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Romana Jarosova
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
- Department of Analytical Chemistry, UNESCO Laboratory of Environmental Electrochemistry, Charles University, Prague 2, Czech Republic 12843
| | - Michael A. Johnson
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
30
|
Wullimann MF. The Neuromeric/Prosomeric Model in Teleost Fish Neurobiology. BRAIN, BEHAVIOR AND EVOLUTION 2022; 97:336-360. [PMID: 35728561 PMCID: PMC9808694 DOI: 10.1159/000525607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 06/08/2022] [Indexed: 01/07/2023]
Abstract
The neuromeric/prosomeric model has been rejuvenated by Puelles and Rubenstein [Trends Neurosci. 1993;16(11):472-9]. Here, its application to the (teleostean) fish brain is detailed, beginning with a historical account. The second part addresses three main issues with particular interest for fish neuroanatomy and looks at the impact of the neuromeric model on their understanding. The first one is the occurrence of four early migrating forebrain areas (M1 through M4) in teleosts and their comparative interpretation. The second issue addresses the complex development and neuroanatomy of the teleostean alar and basal hypothalamus. The third topic is the vertebrate dopaminergic system, with the focus on some teleostean peculiarities. Most of the information will be coming from zebrafish studies, although the general ductus is a comparative one. Throughout the manuscript, comparative developmental and organizational aspects of the teleostean amygdala are discussed. One particular focus is cellular migration streams into the medial amygdala.
Collapse
Affiliation(s)
- Mario F. Wullimann
- Division of Neurobiology, Department Biologie II, Ludwig-Maximilians-Universität München (LMU Munich), Martinsried, Germany,Department Genes-Circuits-Behavior, Max-Planck-Institute for Biological Intelligence (i.F.), Martinsried, Germany,*Mario F. Wullimann,
| |
Collapse
|
31
|
Environmentally-relevant concentrations of the antipsychotic drugs sulpiride and clozapine induce abnormal dopamine and serotonin signaling in zebrafish brain. Sci Rep 2022; 12:17973. [PMID: 36289270 PMCID: PMC9606268 DOI: 10.1038/s41598-022-22169-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
The presence of drugs in surface and groundwaters adversely affects the physiological function of non-target organisms due special activities that can pose a serious threats to various forms of aquatic life. Psychotropic drugs are one of the most commonly used drugs in the world. Hence, the aim of this study was to investigate the effect of environmentally-relevant concentrations of the antipsychotic drugs, sulpiride and clozapine, on dopaminergic (DAergic) and serotonergic (5-HTergic) neurotransmitter systems in the brain of zebrafish. Adult zebrafish (AB strain) were exposed to the environmentally-relevant concentrations of sulpiride, clozapine, or a mixture of sulpiride and clozapine. The effects of the drugs on the mRNA and protein levels of major functional molecules in DAergic and 5-HTergic systems were then analyzed in the telencephalon and diencephalon. Both drugs induced abnormal mRNA and protein levels of important functional molecules of the DA and 5-HT signaling pathways in both telencephalon and diencephalon, as shown by the abnormal transcriptional levels of TH, DAT, DR D1, DR D2, MAO, TPH, serotonin transporter (SERT), 5-HTR 1AA, 5-HTR 1B, 5-THR 2AA, and 5-HTR 2B, and the abnormal translational levels of DAT, DR D2, SERT, 5-HTR 1A, 5-HTR 1B, and 5-HTR 2B. In addition, we observed a specificity in the adverse effects of these antipsychotic drugs, in terms of doses and brain parts. Compared to their effects alone, the drug mixture had a weaker effect on the DA and 5-HT systems, suggesting an antagonistic interaction between sulpiride and clozapine. Our findings suggest that sulpiride and clozapine interfere with DAergic and 5-HTergic neurotransmitter systems in the telencephalon and diencephalon of zebrafish, resulting in possible effects on brain functions and posing a serious threat to the health of zebrafish.
Collapse
|
32
|
Chia K, Klingseisen A, Sieger D, Priller J. Zebrafish as a model organism for neurodegenerative disease. Front Mol Neurosci 2022; 15:940484. [PMID: 36311026 PMCID: PMC9606821 DOI: 10.3389/fnmol.2022.940484] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
The zebrafish is increasingly recognized as a model organism for translational research into human neuropathology. The zebrafish brain exhibits fundamental resemblance with human neuroanatomical and neurochemical pathways, and hallmarks of human brain pathology such as protein aggregation, neuronal degeneration and activation of glial cells, for example, can be modeled and recapitulated in the fish central nervous system. Genetic manipulation, imaging, and drug screening are areas where zebrafish excel with the ease of introducing mutations and transgenes, the expression of fluorescent markers that can be detected in vivo in the transparent larval stages overtime, and simple treatment of large numbers of fish larvae at once followed by automated screening and imaging. In this review, we summarize how zebrafish have successfully been employed to model human neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, and Huntington’s disease. We discuss advantages and disadvantages of choosing zebrafish as a model for these neurodegenerative conditions.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Klingseisen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Dirk Sieger
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Dirk Sieger,
| | - Josef Priller
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, DZNE, Berlin, Germany
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Josef Priller,
| |
Collapse
|
33
|
Hettiarachchi P, Johnson MA. Characterization of D3 Autoreceptor Function in Whole Zebrafish Brain with Fast-Scan Cyclic Voltammetry. ACS Chem Neurosci 2022; 13:2863-2873. [PMID: 36099546 PMCID: PMC10105970 DOI: 10.1021/acschemneuro.2c00280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Zebrafish (Danio rerio) are ideal model organisms for investigating nervous system function, both in health and disease. Nevertheless, functional characteristics of dopamine (DA) release and uptake regulation are still not well-understood in zebrafish. In this study, we assessed D3 autoreceptor function in the telencephalon of whole zebrafish brains ex vivo by measuring the electrically stimulated DA release ([DA]max) and uptake at carbon fiber microelectrodes with fast-scan cyclic voltammetry. Treatment with pramipexole and 7-OH-DPAT, selective D3 autoreceptor agonists, sharply decreased [DA]max. Conversely, SB277011A, a selective D3 antagonist, nearly doubled [DA]max and decreased k, the first-order rate constant for the DA uptake, to about 20% of its original value. Treatment with desipramine, a selective norepinephrine transporter blocker, failed to increase current, suggesting that our electrochemical signal arises solely from the release of DA. Furthermore, blockage of DA uptake with nomifensine-reversed 7-OH-DPAT induced decreases in [DA]max. Collectively, our data show that, as in mammals, D3 autoreceptors regulate DA release, likely by inhibiting uptake. The results of this study are useful in the further development of zebrafish as a model organism for DA-related neurological disorders such as Parkinson's disease, schizophrenia, and drug addiction.
Collapse
Affiliation(s)
- Piyanka Hettiarachchi
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Michael A Johnson
- Department of Chemistry and R.N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
34
|
Son JH, Gerenza AK, Bingener GM, Bonkowsky JL. Hypoplasia of dopaminergic neurons by hypoxia-induced neurotoxicity is associated with disrupted swimming development of larval zebrafish. Front Cell Neurosci 2022; 16:963037. [PMID: 36212692 PMCID: PMC9540391 DOI: 10.3389/fncel.2022.963037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Hypoxic injury to the developing brain increases the risk of permanent behavioral deficits, but the precise mechanisms of hypoxic injury to the developing nervous system are poorly understood. In this study, we characterized the effects of developmental hypoxia (1% pO2 from 24 to 48 h post-fertilization, hpf) on diencephalic dopaminergic (DA) neurons in larval zebrafish and the consequences on the development of swimming behavior. Hypoxia reduced the number of diencephalic DA neurons at 48 hpf. Returning zebrafish larvae to normoxia after the hypoxia (i.e., hypoxia-recovery, HR) induced reactive oxygen species (ROS) accumulation. Real-time qPCR results showed that HR caused upregulation of proapoptotic genes, including p53 and caspase3, suggesting the potential for ROS-induced cell death. With HR, we also found an increase in TUNEL-positive DA neurons, a persistent reduction in the number of diencephalic DA neurons, and disrupted swimming development and behavior. Interestingly, post-hypoxia (HR) with the antioxidant N-acetylcysteine partially restored the number of DA neurons and spontaneous swimming behavior, demonstrating potential recovery from hypoxic injury. The present study provides new insights for understanding the mechanisms responsible for motor disability due to developmental hypoxic injury.
Collapse
Affiliation(s)
- Jong-Hyun Son
- Department of Biology, Neuroscience Program, University of Scranton, Scranton, PA, United States
- *Correspondence: Jong-Hyun Son,
| | - Amanda K. Gerenza
- Department of Biology, Neuroscience Program, University of Scranton, Scranton, PA, United States
| | - Gabrielle M. Bingener
- Department of Biology, Neuroscience Program, University of Scranton, Scranton, PA, United States
| | - Joshua L. Bonkowsky
- Department of Pediatrics, School of Medicine, Brain and Spine Center, Primary Children’s Hospital, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
35
|
Eugenin von Bernhardi J, Biechl D, Miek L, Herget U, Ryu S, Wullimann MF. A versatile transcription factor: Multiple roles of orthopedia a (otpa) beyond its restricted localization in dopaminergic systems of developing and adult zebrafish (Danio rerio) brains. J Comp Neurol 2022; 530:2537-2561. [PMID: 35708548 DOI: 10.1002/cne.25351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/06/2022]
Abstract
Many transcription factors boost neural development and differentiation in specific directions and serve for identifying similar or homologous structures across species. The expression of Orthopedia (Otp) is critical for the development of certain cell groups along the vertebrate neuraxis, for example, the medial amygdala or hypothalamic neurosecretory neurons. Therefore, the primary focus of the present study is the distribution of Orthopedia a (Otpa) in the larval and adult zebrafish (Danio rerio) brain. Since Otpa is also critical for the development of zebrafish basal diencephalic dopaminergic cells, colocalization of Otpa with the catecholamine synthesizing enzyme tyrosine hydroxylase (TH) is studied. Cellular colocalization of Otpa and dopamine is only seen in magnocellular neurons of the periventricular posterior tubercular nucleus and in the posterior tuberal nucleus. Otpa-positive cells occur in many additional structures along the zebrafish neuraxis, from the secondary prosencephalon down to the hindbrain. Furthermore, Otpa expression is studied in shh-GFP and islet1-GFP transgenic zebrafish. Otpa-positive cells only express shh in dopaminergic magnocellular periventricular posterior tubercular cells, and only colocalize with islet1-GFP in the ventral zone and prerecess caudal periventricular hypothalamic zone and the perilemniscal nucleus. The scarcity of cellular colocalization of Otpa in islet1-GFP cells indicates that the Shh-islet1 neurogenetic pathway is not active in most Otpa-expressing domains. Our analysis reveals detailed correspondences between mouse and zebrafish forebrain territories including the zebrafish intermediate nucleus of the ventral telencephalon and the mouse medial amygdala. The zebrafish preoptic Otpa-positive domain represents the neuropeptidergic supraopto-paraventricular region of all tetrapods. Otpa domains in the zebrafish basal plate hypothalamus suggest that the ventral periventricular hypothalamic zone corresponds to the otp-expressing basal hypothalamic tuberal field in the mouse. Furthermore, the mouse otp domain in the mammillary hypothalamus compares partly to our Otpa-positive domain in the prerecess caudal periventricular hypothalamic zone (Hc-a).
Collapse
Affiliation(s)
- Jaime Eugenin von Bernhardi
- Faculty of Biology, Division of Neurobiology, Ludwig-Maximilians-Universität Munich, München, Bavaria, Germany.,The Solomon Snyder Department of Neuroscience, Johns Hopkins Univeristy, Baltimore, Maryland, USA
| | - Daniela Biechl
- Faculty of Biology, Division of Neurobiology, Ludwig-Maximilians-Universität Munich, München, Bavaria, Germany
| | - Laura Miek
- Faculty of Biology, Division of Neurobiology, Ludwig-Maximilians-Universität Munich, München, Bavaria, Germany
| | - Ulrich Herget
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Soojin Ryu
- Living Systems Institute University of Exeter, Exeter, Devon, UK.,College of Medicine and Health, University of Exeter, Exeter, Devon, UK
| | - Mario F Wullimann
- Faculty of Biology, Division of Neurobiology, Ludwig-Maximilians-Universität Munich, München, Bavaria, Germany.,Max-Planck-Institute of Neurobiology, Planegg-Martinsried, Germany
| |
Collapse
|
36
|
Perdikaris P, Dermon CR. Behavioral and neurochemical profile of MK-801 adult zebrafish model: Forebrain β 2-adrenoceptors contribute to social withdrawal and anxiety-like behavior. Prog Neuropsychopharmacol Biol Psychiatry 2022; 115:110494. [PMID: 34896197 DOI: 10.1016/j.pnpbp.2021.110494] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/22/2021] [Accepted: 12/03/2021] [Indexed: 01/29/2023]
Abstract
Deficits in social communication and interaction are core clinical symptoms characterizing multiple neuropsychiatric conditions, including autism spectrum disorder (ASD) and schizophrenia. Interestingly, elevated anxiety levels are a common comorbid psychopathology characterizing individuals with aberrant social behavior. Despite recent progress, the underlying neurobiological mechanisms that link anxiety with social withdrawal remain poorly understood. The present study developed a zebrafish pharmacological model displaying social withdrawal behavior, following a 3-h exposure to 4 μΜ (+)-MK-801, a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, for 7 days. Interestingly, MK-801-treated zebrafish displayed elevated anxiety levels along with higher frequency of stereotypical behaviors, rendering this zebrafish model appropriate to unravel a possible link of catecholaminergic and ASD-like phenotypes. MK-801-treated zebrafish showed increased telencephalic protein expression of metabotropic glutamate 5 receptor (mGluR5), dopamine transporter (DAT) and β2-adrenergic receptors (β2-ARs), supporting the presence of excitation/inhibition imbalance along with altered dopaminergic and noradrenergic activity. Interestingly, β2-ARs expression, was differentially regulated across the Social Decision-Making (SDM) network nodes, exhibiting increased levels in ventral telencephalic area (Vv), a key-area integrating reward and social circuits but decreased expression in dorso-medial telencephalic area (Dm) and anterior tuberal nucleus (ATN). Moreover, the co-localization of β2-ARs with elements of GABAergic and glutamatergic systems, as well as with GAP-43, a protein indicating increased brain plasticity potential, support the key-role of β2-ARs in the MK-801 zebrafish social dysfunctions. Our results highlight the importance of the catecholaminergic neurotransmission in the manifestation of ASD-like behavior, representing a site of potential interventions for amelioration of ASD-like symptoms.
Collapse
Affiliation(s)
- Panagiotis Perdikaris
- Human and Animal Physiology Laboratory, Department of Biology, University of Patras, Rio, 26500 Patras, Greece
| | - Catherine R Dermon
- Human and Animal Physiology Laboratory, Department of Biology, University of Patras, Rio, 26500 Patras, Greece.
| |
Collapse
|
37
|
Gundlach M, Di Paolo C, Chen Q, Majewski K, Haigis AC, Werner I, Hollert H. Clozapine modulation of zebrafish swimming behavior and gene expression as a case study to investigate effects of atypical drugs on aquatic organisms. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 815:152621. [PMID: 34968598 DOI: 10.1016/j.scitotenv.2021.152621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/01/2021] [Accepted: 12/19/2021] [Indexed: 06/14/2023]
Abstract
Mental illnesses affect more than 150 million people in Europe and lead to an increasing consumption of neuroactive drugs during the last twenty years. The antipsychotic compound, clozapine, is one of the most used psychotropic drugs worldwide, with potentially negative consequences for the aquatic environment. Hence, the objectives of the study presented here were the quantification of clozapine induced changes in swimming behavior of exposed Danio rerio embryos and the elucidation of the molecular effects on the serotonergic and dopaminergic systems. Yolk-sac larvae were exposed to different concentrations (0.2 mg/L, 0.4 mg/L, 0.8 mg/L, 1.6 mg/L, 3.2 mg/L and 6.4 mg/L) of clozapine for 116 h post-fertilization, and changes in the swimming behavior of the larvae were assessed. Further, quantitative real-time PCR was performed to analyze the expression of selected genes. The qualitative evaluation of changes in the swimming behavior of D. rerio larvae revealed a significant decrease of the average swimming distance and velocity in the light-dark transition test, with more than a 36% reduction at the highest exposure concentration of 6.4 mg/L. Furthermore, the total larval body length was reduced at the highest concentration. An in-depth analysis based on expression of selected target genes of the serotonin (slc6a4a) and dopamine (drd2a) system showed an upregulation at a concentration of 1.6 mg/L and above. In addition, a lower increase in expression was detected for biomarkers of general stress (adra1a and cyp1a2). Our data show that exposure to clozapine during development inhibits swimming activity of zebrafish larvae, which could, in part, be due to disruption of the serotonin- and dopamine system.
Collapse
Affiliation(s)
- Michael Gundlach
- Department of Ecosystem Analysis, Institute for Environmental Research, ABBt-Aachen Biology and Biotechnology, RWTH Aachen University, 52074 Aachen, Germany
| | - Carolina Di Paolo
- Department of Ecosystem Analysis, Institute for Environmental Research, ABBt-Aachen Biology and Biotechnology, RWTH Aachen University, 52074 Aachen, Germany
| | - Qiqing Chen
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Kendra Majewski
- Department of Ecosystem Analysis, Institute for Environmental Research, ABBt-Aachen Biology and Biotechnology, RWTH Aachen University, 52074 Aachen, Germany
| | - Ann-Cathrin Haigis
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Inge Werner
- Swiss Centre for Applied Ecotoxicology, Überlandstrasse 131, 8600 Dübendorf, Switzerland
| | - Henner Hollert
- Department of Ecosystem Analysis, Institute for Environmental Research, ABBt-Aachen Biology and Biotechnology, RWTH Aachen University, 52074 Aachen, Germany; Department Evolutionary Ecology and Environmental Toxicology, Faculty Biological Sciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
38
|
Dwortz MF, Curley JP, Tye KM, Padilla-Coreano N. Neural systems that facilitate the representation of social rank. Philos Trans R Soc Lond B Biol Sci 2022; 377:20200444. [PMID: 35000438 PMCID: PMC8743891 DOI: 10.1098/rstb.2020.0444] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Across species, animals organize into social dominance hierarchies that serve to decrease aggression and facilitate survival of the group. Neuroscientists have adopted several model organisms to study dominance hierarchies in the laboratory setting, including fish, reptiles, rodents and primates. We review recent literature across species that sheds light onto how the brain represents social rank to guide socially appropriate behaviour within a dominance hierarchy. First, we discuss how the brain responds to social status signals. Then, we discuss social approach and avoidance learning mechanisms that we propose could drive rank-appropriate behaviour. Lastly, we discuss how the brain represents memories of individuals (social memory) and how this may support the maintenance of unique individual relationships within a social group. This article is part of the theme issue 'The centennial of the pecking order: current state and future prospects for the study of dominance hierarchies'.
Collapse
Affiliation(s)
- Madeleine F. Dwortz
- Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - James P. Curley
- Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
| | - Kay M. Tye
- Systems Neuroscience Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Nancy Padilla-Coreano
- Systems Neuroscience Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Department of Neuroscience, University of Florida, Gainesville, FN 32611, USA
| |
Collapse
|
39
|
Ogawa S, Parhar IS. Role of Habenula in Social and Reproductive Behaviors in Fish: Comparison With Mammals. Front Behav Neurosci 2022; 15:818782. [PMID: 35221943 PMCID: PMC8867168 DOI: 10.3389/fnbeh.2021.818782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/27/2021] [Indexed: 02/05/2023] Open
Abstract
Social behaviors such as mating, parenting, fighting, and avoiding are essential functions as a communication tool in social animals, and are critical for the survival of individuals and species. Social behaviors are controlled by a complex circuitry that comprises several key social brain regions, which is called the social behavior network (SBN). The SBN further integrates social information with external and internal factors to select appropriate behavioral responses to social circumstances, called social decision-making. The social decision-making network (SDMN) and SBN are structurally, neurochemically and functionally conserved in vertebrates. The social decision-making process is also closely influenced by emotional assessment. The habenula has recently been recognized as a crucial center for emotion-associated adaptation behaviors. Here we review the potential role of the habenula in social function with a special emphasis on fish studies. Further, based on evolutional, molecular, morphological, and behavioral perspectives, we discuss the crucial role of the habenula in the vertebrate SDMN.
Collapse
|
40
|
Bashirzade AAO, Cheresiz SV, Belova AS, Drobkov AV, Korotaeva AD, Azizi-Arani S, Azimirad A, Odle E, Gild EYV, Ardashov OV, Volcho KP, Bozhko DV, Myrov VO, Kolchanova SM, Polovian AI, Galumov GK, Salakhutdinov NF, Amstislavskaya TG, Kalueff AV. MPTP-Treated Zebrafish Recapitulate 'Late-Stage' Parkinson's-like Cognitive Decline. TOXICS 2022; 10:69. [PMID: 35202255 PMCID: PMC8879925 DOI: 10.3390/toxics10020069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/25/2022]
Abstract
The zebrafish is a promising model species in biomedical research, including neurotoxicology and neuroactive drug screening. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) evokes degeneration of dopaminergic neurons and is commonly used to model Parkinson's disease (PD) in laboratory animals, including zebrafish. However, cognitive phenotypes in MPTP-evoked experimental PD models remain poorly understood. Here, we established an LD50 (292 mg/kg) for intraperitoneal MPTP administration in adult zebrafish, and report impaired spatial working memory (poorer spontaneous alternation in the Y-maze) in a PD model utilizing fish treated with 200 µg of this agent. In addition to conventional behavioral analyses, we also employed artificial intelligence (AI)-based approaches to independently and without bias characterize MPTP effects on zebrafish behavior during the Y-maze test. These analyses yielded a distinct cluster for 200-μg MPTP (vs. other) groups, suggesting that high-dose MPTP produced distinct, computationally detectable patterns of zebrafish swimming. Collectively, these findings support MPTP treatment in adult zebrafish as a late-stage experimental PD model with overt cognitive phenotypes.
Collapse
Affiliation(s)
- Alim A. O. Bashirzade
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Sergey V. Cheresiz
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Alisa S. Belova
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Alexey V. Drobkov
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Anastasiia D. Korotaeva
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Soheil Azizi-Arani
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Amirhossein Azimirad
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Eric Odle
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Emma-Yanina V. Gild
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Oleg V. Ardashov
- Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia; (O.V.A.); (K.P.V.); (N.F.S.)
| | - Konstantin P. Volcho
- Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia; (O.V.A.); (K.P.V.); (N.F.S.)
| | - Dmitrii V. Bozhko
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | - Vladislav O. Myrov
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | - Sofia M. Kolchanova
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | | | - Georgii K. Galumov
- ZebraML, Inc., Houston, TX 77043, USA; (D.V.B.); (V.O.M.); (S.M.K.); (A.I.P.); (G.K.G.)
| | - Nariman F. Salakhutdinov
- Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 630090 Novosibirsk, Russia; (O.V.A.); (K.P.V.); (N.F.S.)
| | - Tamara G. Amstislavskaya
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
| | - Allan V. Kalueff
- Scientific Research Institute of Neuroscience and Medicine, 630090 Novosibirsk, Russia; (S.V.C.); (A.S.B.); (T.G.A.)
- Institute of Medicine and Psychology, Novosibirsk State University, 630117 Novosibirsk, Russia; (A.V.D.); (A.D.K.); (S.A.-A.); (A.A.); (E.O.); (E.-Y.V.G.)
- Ural Federal University, 620002 Yekaterinburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, 354340 Sochi, Russia
- Moscow Institute of Physics and Technology, 141701 Moscow, Russia
- Granov Scientific Research Center of Radiology and Surgical Technologies, 197758 St. Petersburg, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia
- School of Pharmacy, Southwest University, Chongqing 400715, China
| |
Collapse
|
41
|
Ghahramani ZN, Perelmuter JT, Varughese J, Kyaw P, Palmer WC, Sisneros JA, Forlano PM. Activation of noradrenergic locus coeruleus and social behavior network nuclei varies with duration of male midshipman advertisement calls. Behav Brain Res 2022; 423:113745. [PMID: 35033611 DOI: 10.1016/j.bbr.2022.113745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Vocal courtship is vital to the reproductive success of many vertebrates and is therefore a highly-motivated behavioral state. Catecholamines have been shown to play an essential role in the expression and maintenance of motivated vocal behavior, such as the coordination of vocal-motor output in songbirds. However, it is not well-understood if this relationship applies to anamniote vocal species. Using the plainfin midshipman fish model, we tested whether specific catecholaminergic (i.e., dopaminergic and noradrenergic) nuclei and nodes of the social behavior network (SBN) are differentially activated in vocally courting (humming) versus non-humming males. Herein, we demonstrate that tyrosine hydroxylase immunoreactive (TH-ir) neuron number in the noradrenergic locus coeruleus (LC) and induction of cFos (an immediate early gene product and proxy for neural activation) in the preoptic area differentiated humming from non-humming males. Furthermore, we found relationships between activation of the LC and SBN nuclei with the total amount of time that males spent humming, further reinforcing a role for these specific brain regions in the production of motivated reproductive-related vocalizations. Finally, we found that patterns of functional connectivity between catecholaminergic nuclei and nodes of the SBN differed between humming and non-humming males, supporting the notion that adaptive behaviors (such as the expression of advertisement hums) emerge from the interactions between various catecholaminergic nuclei and the SBN.
Collapse
Affiliation(s)
- Zachary N Ghahramani
- Department of Biological Sciences, University of Mary Washington, Fredericksburg, VA, USA; Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY, USA; Doctoral Subprograms in Ecology, Evolutionary Biology and Behavior,.
| | - Jonathan T Perelmuter
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA; Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY, USA; Neuroscience, and Behavioral and Cognitive Neuroscience, The Graduate Center, City University of New York, New York, NY, USA
| | - Joshua Varughese
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY, USA
| | - Phoo Kyaw
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY, USA
| | | | - Joseph A Sisneros
- Departments of Biology and Psychology,; University of Washington, Seattle, WA, USA; Virginia Bloedel Hearing Research Center, Seattle, WA, USA
| | - Paul M Forlano
- Department of Biology, Brooklyn College, City University of New York, Brooklyn, NY, USA; Doctoral Subprograms in Ecology, Evolutionary Biology and Behavior,; Neuroscience, and Behavioral and Cognitive Neuroscience, The Graduate Center, City University of New York, New York, NY, USA; Behavioral and Cognitive Neuroscience, The Graduate Center, City University of New York, New York, NY, USA.
| |
Collapse
|
42
|
Crouzier L, Richard EM, Sourbron J, Lagae L, Maurice T, Delprat B. Use of Zebrafish Models to Boost Research in Rare Genetic Diseases. Int J Mol Sci 2021; 22:13356. [PMID: 34948153 PMCID: PMC8706563 DOI: 10.3390/ijms222413356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Rare genetic diseases are a group of pathologies with often unmet clinical needs. Even if rare by a single genetic disease (from 1/2000 to 1/more than 1,000,000), the total number of patients concerned account for approximatively 400 million peoples worldwide. Finding treatments remains challenging due to the complexity of these diseases, the small number of patients and the challenge in conducting clinical trials. Therefore, innovative preclinical research strategies are required. The zebrafish has emerged as a powerful animal model for investigating rare diseases. Zebrafish combines conserved vertebrate characteristics with high rate of breeding, limited housing requirements and low costs. More than 84% of human genes responsible for diseases present an orthologue, suggesting that the majority of genetic diseases could be modelized in zebrafish. In this review, we emphasize the unique advantages of zebrafish models over other in vivo models, particularly underlining the high throughput phenotypic capacity for therapeutic screening. We briefly introduce how the generation of zebrafish transgenic lines by gene-modulating technologies can be used to model rare genetic diseases. Then, we describe how zebrafish could be phenotyped using state-of-the-art technologies. Two prototypic examples of rare diseases illustrate how zebrafish models could play a critical role in deciphering the underlying mechanisms of rare genetic diseases and their use to identify innovative therapeutic solutions.
Collapse
Affiliation(s)
- Lucie Crouzier
- MMDN, University of Montpellier, EPHE, INSERM, 34095 Montpellier, France; (L.C.); (E.M.R.); (T.M.)
| | - Elodie M. Richard
- MMDN, University of Montpellier, EPHE, INSERM, 34095 Montpellier, France; (L.C.); (E.M.R.); (T.M.)
| | - Jo Sourbron
- Department of Development and Regeneration, Section Pediatric Neurology, University Hospital KU Leuven, 3000 Leuven, Belgium; (J.S.); (L.L.)
| | - Lieven Lagae
- Department of Development and Regeneration, Section Pediatric Neurology, University Hospital KU Leuven, 3000 Leuven, Belgium; (J.S.); (L.L.)
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, 34095 Montpellier, France; (L.C.); (E.M.R.); (T.M.)
| | - Benjamin Delprat
- MMDN, University of Montpellier, EPHE, INSERM, 34095 Montpellier, France; (L.C.); (E.M.R.); (T.M.)
| |
Collapse
|
43
|
Kenney JW, Steadman PE, Young O, Shi MT, Polanco M, Dubaishi S, Covert K, Mueller T, Frankland PW. A 3D adult zebrafish brain atlas (AZBA) for the digital age. eLife 2021; 10:69988. [PMID: 34806976 PMCID: PMC8639146 DOI: 10.7554/elife.69988] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 11/21/2021] [Indexed: 01/19/2023] Open
Abstract
Zebrafish have made significant contributions to our understanding of the vertebrate brain and the neural basis of behavior, earning a place as one of the most widely used model organisms in neuroscience. Their appeal arises from the marriage of low cost, early life transparency, and ease of genetic manipulation with a behavioral repertoire that becomes more sophisticated as animals transition from larvae to adults. To further enhance the use of adult zebrafish, we created the first fully segmented three-dimensional digital adult zebrafish brain atlas (AZBA). AZBA was built by combining tissue clearing, light-sheet fluorescence microscopy, and three-dimensional image registration of nuclear and antibody stains. These images were used to guide segmentation of the atlas into over 200 neuroanatomical regions comprising the entirety of the adult zebrafish brain. As an open source, online (azba.wayne.edu), updatable digital resource, AZBA will significantly enhance the use of adult zebrafish in furthering our understanding of vertebrate brain function in both health and disease.
Collapse
Affiliation(s)
- Justin W Kenney
- Department of Biological Sciences, Wayne State University, Detroit, United States.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
| | - Patrick E Steadman
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
| | - Olivia Young
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Meng Ting Shi
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Maris Polanco
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Saba Dubaishi
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Kristopher Covert
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Thomas Mueller
- Division of Biology, Kansas State University, Manhattan, United States
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Canada.,Department of Psychology, University of Toronto, Toronto, Canada
| |
Collapse
|
44
|
Clayman CL, Connaughton VP. Neurochemical and Behavioral Consequences of Ethanol and/or Caffeine Exposure: Effects in Zebrafish and Rodents. Curr Neuropharmacol 2021; 20:560-578. [PMID: 34766897 DOI: 10.2174/1570159x19666211111142027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022] Open
Abstract
Zebrafish are increasingly being utilized to model the behavioral and neurochemical effects of pharmaceuticals and, more recently, pharmaceutical interactions. Zebrafish models of stress establish that both caffeine and ethanol influence anxiety, though few studies have implemented co-administration to assess the interaction of anxiety and reward-seeking. Caffeine exposure in zebrafish is teratogenic, causing developmental abnormalities in the cardiovascular, neuromuscular, and nervous systems of embryos and larvae. Ethanol is also a teratogen and, as an anxiolytic substance, may be able to offset the anxiogenic effects of caffeine. Co-exposure to caffeine and alcohol impacts neuroanatomy and behavior in adolescent animal models, suggesting stimulant substances may moderate the impact of alcohol on neural circuit development. Here, we review the literature describing neuropharmacological and behavioral consequences of caffeine and/or alcohol exposure in the zebrafish model, focusing on neurochemistry, locomotor effects, and behavioral assessments of stress/anxiety as reported in adolescent/juvenile and adult animals. The purpose of this review is twofold: (1) describe the work in zebrafish documenting the effects of ethanol and/or caffeine exposure and (2) compare these zebrafish studies with comparable experiments in rodents. We focus on specific neurochemical pathways (dopamine, serotonin, adenosine, GABA, adenosine), anxiety-type behaviors (assessed with novel tank, thigmotaxis, shoaling), and locomotor changes resulting from both individual and co-exposure. We compare findings in zebrafish with those in rodent models, revealing similarities across species and identifying conservation of mechanisms that potentially reinforce co-addiction.
Collapse
Affiliation(s)
- Carly L Clayman
- Department of Biology and Center for Neuroscience and Behavior American University, Washington, DC 20016, United States
| | - Victoria P Connaughton
- Department of Biology and Center for Neuroscience and Behavior American University, Washington, DC 20016, United States
| |
Collapse
|
45
|
Natterson-Horowitz B, Cho JH. Stress, Subordination, and Anomalies of Feeding Across the Tree of Life: Implications for Interpreting Human Eating Disorders. Front Psychol 2021; 12:727554. [PMID: 34675841 PMCID: PMC8525799 DOI: 10.3389/fpsyg.2021.727554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Eating behaviors of animals living in naturalistic environments offer unique insights into several dysregulated eating patterns observed in humans. Social subordination is a known precipitant of hyperphagia and hypophagia in human beings, and examples of similar responses have been identified in a phylogenetically widespread range of vertebral species. This points to potentially conserved, patterned responses to animals navigating lives within social hierarchies. Self-imposed food restriction in subordinate fish and hyperphagic responses in socially subordinated bird and primate individuals may represent evolved adaptations to the stress of social subordination. As such, hyperphagic and hypophagic responses to social subordination in these species may model the natural history, neurobiology, and behavioral ecology of human dieting and bingeing more accurately than some current animal models. Phylogenetically widespread similarities in eating patterns under the stress of social subordination point to potentially shared biological benefits of these behaviors across species and the role of evolutionary trade-offs, adaptations, and other processes in shaping them. The application of a broadly comparative lens to disordered eating behaviors in other species exposes important similarities and differences between neurophysiology of eating across species. In doing so, it highlights the value of phylogenetic analyses and macroevolution as tools for identifying novel, naturally occurring models for understanding disordered human eating. Moreover, this approach introduces the intriguing possibility that human cultural influences on disordered eating may have far more ancient origins than previously considered.
Collapse
Affiliation(s)
- B Natterson-Horowitz
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Department of Human Evolutionary Biology, Harvard University, Boston, MA, United States
| | - Julia H Cho
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
46
|
Abstract
The olfactory system allows animals to navigate in their environment to feed, mate, and escape predators. It is well established that odorant exposure or electrical stimulation of the olfactory system induces stereotyped motor responses in fishes. However, the neural circuitry responsible for the olfactomotor transformations is only beginning to be unraveled. A neural substrate eliciting motor responses to olfactory inputs was identified in the lamprey, a basal vertebrate used extensively to examine the neural mechanisms underlying sensorimotor transformations. Two pathways were discovered from the olfactory organ in the periphery to the brainstem motor nuclei responsible for controlling swimming. The first pathway originates from sensory neurons located in the accessory olfactory organ and reaches a single population of projection neurons in the medial olfactory bulb, which, in turn, transmit the olfactory signals to the posterior tuberculum and then to downstream brainstem locomotor centers. A second pathway originates from the main olfactory epithelium and reaches the main olfactory bulb, the neurons of which project to the pallium/cortex. The olfactory signals are then conveyed to the posterior tuberculum and then to brainstem locomotor centers. Olfactomotor behavior can adapt, and studies were aimed at defining the underlying neural mechanisms. Modulation of bulbar neural activity by GABAergic, dopaminergic, and serotoninergic inputs is likely to provide strong control over the hardwired circuits to produce appropriate motor behavior in response to olfactory cues. This review summarizes current knowledge relative to the neural circuitry producing olfactomotor behavior in lampreys and their modulatory mechanisms.
Collapse
|
47
|
Nunes AR, Gliksberg M, Varela SAM, Teles M, Wircer E, Blechman J, Petri G, Levkowitz G, Oliveira RF. Developmental Effects of Oxytocin Neurons on Social Affiliation and Processing of Social Information. J Neurosci 2021; 41:8742-8760. [PMID: 34470805 PMCID: PMC8528494 DOI: 10.1523/jneurosci.2939-20.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 11/21/2022] Open
Abstract
Hormones regulate behavior either through activational effects that facilitate the acute expression of specific behaviors or through organizational effects that shape the development of the nervous system thereby altering adult behavior. Much research has implicated the neuropeptide oxytocin (OXT) in acute modulation of various aspects of social behaviors across vertebrate species, and OXT signaling is associated with the developmental social deficits observed in autism spectrum disorders (ASDs); however, little is known about the role of OXT in the neurodevelopment of the social brain. We show that perturbation of OXT neurons during early zebrafish development led to a loss of dopaminergic neurons, associated with visual processing and reward, and blunted the neuronal response to social stimuli in the adult brain. Ultimately, adult fish whose OXT neurons were ablated in early life, displayed altered functional connectivity within social decision-making brain nuclei both in naive state and in response to social stimulus and became less social. We propose that OXT neurons have an organizational role, namely, to shape forebrain neuroarchitecture during development and to acquire an affiliative response toward conspecifics.SIGNIFICANCE STATEMENT Social behavior is developed over the lifetime of an organism and the neuropeptide oxytocin (OXT) modulates social behaviors across vertebrate species, and is associated with neuro-developmental social deficits such as autism. However, whether OXT plays a role in the developmental maturation of neural systems that are necessary for social behavior remains poorly explored. We show that proper behavioral and neural response to social stimuli depends on a developmental process orchestrated by OXT neurons. Animals whose OXT system is ablated in early life show blunted neuronal and behavioral responses to social stimuli as well as wide ranging disruptions in the functional connectivity of the social brain. We provide a window into the mechanisms underlying OXT-dependent developmental processes that implement adult sociality.
Collapse
Affiliation(s)
- Ana Rita Nunes
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Susana A M Varela
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- ISPA-Instituto Universitário, Lisboa 1149-041, Portugal
| | - Magda Teles
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Einav Wircer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Giovanni Petri
- Institute for Scientific Interchange (ISI) Foundation and ISI Global Science Foundation, Torino 10126, Italy
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rui F Oliveira
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- ISPA-Instituto Universitário, Lisboa 1149-041, Portugal
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal
| |
Collapse
|
48
|
Fasano G, Godoy RS, Angiulli E, Consalvo A, Franco C, Mancini M, Santucci D, Alleva E, Ciavardelli D, Toni M, Biffali E, Ekker M, Canzoniero LMT, Sordino P. Effects of low-dose methylcyclopentadienyl manganese tricarbonyl-derived manganese on the development of diencephalic dopaminergic neurons in zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117151. [PMID: 34020261 DOI: 10.1016/j.envpol.2021.117151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 04/02/2021] [Accepted: 04/11/2021] [Indexed: 06/12/2023]
Abstract
Fuel additive methylcyclopentadienyl manganese tricarbonyl (MMT) is counted as an organic manganese (Mn)-derived compound. The toxic effects of Mn (alone and complexed) on dopaminergic (DA) neurotransmission have been investigated in both cellular and animal models. However, the impact of environmentally relevant Mn exposure on DA neurodevelopment is rather poorly understood. In the present study, the MMT dose of 100 μM (about 5 mg Mn/L) caused up-regulation of DA-related genes in association with cell body swelling and increase in the number of DA neurons of the ventral diencephalon subpopulation DC2. Furthermore, our analysis identified significant brain Mn bioaccumulation and enhancement of total dopamine levels in association with locomotor hyperactivity. Although DA levels were restored at adulthood, we observed a deficit in the acquisition and consolidation of memory. Collectively, these findings suggest that developmental exposure to low-level MMT-derived Mn is responsible for the selective alteration of diencephalic DA neurons and with long-lasting effects on fish explorative behaviour in adulthood.
Collapse
Affiliation(s)
- Giulia Fasano
- Department of Sciences and Technologies, University of Sannio, Via Francesco de Sanctis, 82100, Benevento, Italy; Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121, Naples, Italy
| | - Rafael Soares Godoy
- Department of Biology, University of Ottawa, Marie-Curie Private, Ottawa, ON K1N 9A7, Canada
| | - Elisa Angiulli
- Department of Biology and Biotechnology ''Charles Darwin", Sapienza University, Via Borelli 50, 00161, Rome, Italy
| | - Ada Consalvo
- Centro Scienze Dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Polacchi 11, 66100, Chieti, Italy; Department of Medical, Oral and Biotechnological Sciences, "G. D'Annunzio" University of Chieti-Pescara, Via Dei Vestini, 66100, Chieti, Italy
| | - Cristina Franco
- Department of Sciences and Technologies, University of Sannio, Via Francesco de Sanctis, 82100, Benevento, Italy
| | - Maria Mancini
- Department of Neuroscience and Physiology, New York University School of Medicine, 435 East 30th Street, New York, NY, 10016, USA; NYU Marlene and Paolo Fresco Institute for Parkinson's Disease and Movement Disorders, New York University School of Medicine, 222 East 41st Street, New York, NY, 10017, USA
| | - Daniela Santucci
- Centro di Riferimento per le Scienze Comportamentali e La Salute Mentale, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Enrico Alleva
- Centro di Riferimento per le Scienze Comportamentali e La Salute Mentale, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Domenico Ciavardelli
- Centro Scienze Dell'Invecchiamento e Medicina Traslazionale - CeSI-MeT, Via Polacchi 11, 66100, Chieti, Italy; School of Human and Social Science, "Kore" University of Enna, Cittadella Universitaria, 94100, Enna, Italy
| | - Mattia Toni
- Department of Biology and Biotechnology ''Charles Darwin", Sapienza University, Via Borelli 50, 00161, Rome, Italy
| | - Elio Biffali
- Department of Research Infrastructures for Marine Biological Resources, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121, Naples, Italy
| | - Marc Ekker
- Department of Biology, University of Ottawa, Marie-Curie Private, Ottawa, ON K1N 9A7, Canada
| | | | - Paolo Sordino
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121, Naples, Italy.
| |
Collapse
|
49
|
Nabinger DD, Altenhofen S, Peixoto JV, da Silva JMK, Bonan CD. Long-lasting behavioral effects of quinpirole exposure on zebrafish. Neurotoxicol Teratol 2021; 88:107034. [PMID: 34600099 DOI: 10.1016/j.ntt.2021.107034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 01/11/2023]
Abstract
The human brain matures into a complex structure, and to reach its complete development, connections must occur along exact paths. If at any stage, the processes are altered, interrupted, or inhibited, the consequences can be permanent. Dopaminergic signaling participates in the control of physiological functions and behavioral processes, and alterations in this signaling pathway are related to the pathogenesis of several neurological disorders. For this reason, the use of pharmacological agents able to interact with the dopaminergic signaling may elucidate the biological bases of such disorders. We investigated the long-lasting behavioral effects on adult zebrafish after quinpirole (a dopamine D2/D3 receptor agonist) exposure during early life stages of development (24 h exposure at 5 days post-fertilization, dpf) to better understand the mechanisms underlying neurological disorders related to the dopaminergic system. Quinpirole exposure at the early life stages of zebrafish led to late behavioral alterations. When evaluated at 120 dpf, zebrafish presented increased anxiety-like behaviors. At the open tank test, fish remained longer at the bottom of the tank, indicating anxiety-like behavior. Furthermore, quinpirole-treated fish exhibited increased absolute turn angle, likely an indication of elevated erratic movements and a sign of increased fear or anxiety. Quinpirole-treated fish also showed altered swimming patterns, characterized by stereotypic swimming. During the open tank test, exposed zebrafish swims from corner to corner in a repetitive manner at the bottom of the tank. Moreover, quinpirole exposure led to memory impairment compared to control fish. However, quinpirole administration had no effects on social and aggressive behavior. These findings demonstrate that dopaminergic signaling altered by quinpirole administration in the early life stages of development led to late alterations in behavioral parameters of adult zebrafish.
Collapse
Affiliation(s)
- Debora Dreher Nabinger
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Stefani Altenhofen
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Julia Vasconcellos Peixoto
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Julia Maria Kuhl da Silva
- Laboratório de Neuroquímica e Psicofarmacologia, Programa de Pós-Graduação em Biologia Celular e 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 Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Cerebrais, Excitotoxicidade e Neuroproteção, Porto Alegre, RS, Brazil.
| |
Collapse
|
50
|
Borgonovo J, Ahumada-Galleguillos P, Oñate-Ponce A, Allende-Castro C, Henny P, Concha ML. Organization of the Catecholaminergic System in the Short-Lived Fish Nothobranchius furzeri. Front Neuroanat 2021; 15:728720. [PMID: 34588961 PMCID: PMC8473916 DOI: 10.3389/fnana.2021.728720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/13/2021] [Indexed: 12/02/2022] Open
Abstract
The catecholaminergic system has received much attention based on its regulatory role in a wide range of brain functions and its relevance in aging and neurodegenerative diseases. In the present study, we analyzed the neuroanatomical distribution of catecholaminergic neurons based on tyrosine hydroxylase (TH) immunoreactivity in the brain of adult Nothobranchius furzeri. In the telencephalon, numerous TH+ neurons were observed in the olfactory bulbs and the ventral telencephalic area, arranged as strips extending through the rostrocaudal axis. We found the largest TH+ groups in the diencephalon at the preoptic region level, the ventral thalamus, the pretectal region, the posterior tuberculum, and the caudal hypothalamus. In the dorsal mesencephalic tegmentum, we identified a particular catecholaminergic group. The rostral rhombencephalon housed TH+ cells in the locus coeruleus and the medulla oblongata, distributing in a region dorsal to the inferior reticular formation, the vagal lobe, and the area postrema. Finally, scattered TH+ neurons were present in the ventral spinal cord and the retina. From a comparative perspective, the overall organization of catecholaminergic neurons is consistent with the general pattern reported for other teleosts. However, N. furzeri shows some particular features, including the presence of catecholaminergic cells in the midbrain. This work provides a detailed neuroanatomical map of the catecholaminergic system of N. furzeri, a powerful aging model, also contributing to the phylogenetic understanding of one of the most ancient neurochemical systems.
Collapse
Affiliation(s)
- Janina Borgonovo
- Laboratory of Experimental Ontogeny, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Patricio Ahumada-Galleguillos
- Laboratory of Experimental Ontogeny, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile
| | - Alejandro Oñate-Ponce
- Laboratory of Experimental Ontogeny, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Camilo Allende-Castro
- Laboratory of Experimental Ontogeny, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Pablo Henny
- Department of Anatomy and Interdisciplinary Center of Neurosciences, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel L Concha
- Laboratory of Experimental Ontogeny, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| |
Collapse
|