1
|
Niaz K, McAtee D, Adhikari P, Rollefson P, Ateia M, Abdelmoneim A. Assessing the effects of fluorine-free and PFAS-containing firefighting foams on development and behavioral responses using a zebrafish-based platform. CHEMOSPHERE 2024; 365:143361. [PMID: 39303789 DOI: 10.1016/j.chemosphere.2024.143361] [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: 06/30/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Significant progress has been made in developing fluorine-free firefighting foams (F3) as alternatives to perfluoroalkyl substances (PFAS)-containing aqueous film-forming foams (AFFF) to help eliminate the health and environmental concerns linked to PFAS exposure. However, developing viable F3 options hinges on a thorough assessment of potential risks alongside the technical performance evaluations. This study showcases the capability of a zebrafish-based platform to discern the developmental and behavioral toxicities associated with exposure to one AFFF and two F3 formulations. To facilitate direct exposure to the chemicals, embryos were enzymatically dechorionated and then exposed to the diluted formulations (6-120 hours post fertilization (hpf)) at concentrations folding from 0.1% of the manufacturer-recommended working concentrations. The exposure regimen also included daily automated media changes (50%) and mortality assessments (24 and 120 hpf). At 120 hpf, a comprehensive assessment encompassing overall development, prevalence of morphological defects, and behavioral responses to acute stressors (visual, acoustic, and peripheral irritant) was conducted. Exposure to both F3s significantly increased larval mortalities to percentages exceeding 90%, whereas AFFF exposures did not cause any significant effect. Overall development, marked by total larval length, was significantly impacted following exposures to all foams. Behavioral responses to acute stressors were also significantly altered following exposures to both F3s, whereas the AFFF did not alter behavior at the concentrations tested. Our findings demonstrate toxicities associated with tested F3 formulations that encompass several endpoints and highlight the utility of the proposed platform in evaluating the developmental toxicities of current and future foam formulations.
Collapse
Affiliation(s)
- Kamal Niaz
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA; Department of Pharmacology and Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 63100, Pakistan
| | - Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Pranup Adhikari
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Patrik Rollefson
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Mohamed Ateia
- United States Environmental Protection Agency, Center for Environmental Solutions & Emergency Response, Cincinnati, OH, 45220, USA.
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
2
|
McAtee D, Abdelmoneim A. Effects of developmental exposure to arsenic species on behavioral stress responses in larval zebrafish and implications for stress-related disorders. Toxicol Sci 2024; 201:61-72. [PMID: 38833692 DOI: 10.1093/toxsci/kfae074] [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: 06/06/2024] Open
Abstract
Arsenic (As) is globally detected in drinking water and food products at levels repeatedly surpassing regulatory thresholds. Several neurological and mental health risks linked to arsenic exposure are proposed; however, the nature of these effects and their association with the chemical forms of arsenic are not fully understood. Gaining a clear understanding of the etiologies and characteristics of these effects is crucial, particularly in association with developmental exposures where the nervous system is most vulnerable. In this study, we investigated the effects of early developmental exposure (6- to 120-h postfertilization [hpf]) of larval zebrafish to environmentally relevant concentrations of arsenic species-trivalent/pentavalent, inorganic/organic forms-on developmental, behavioral, and molecular endpoints to determine their effect on stress response and their potential association with stress-related disorders. At 120 hpf, the developing larvae were assessed for a battery of endpoints including survival, developmental malformities, background activity, and behavioral responses to acute visual and acoustic stimuli. Pooled larval samples were analyzed for alterations in the transcript levels of genes associated with developmental neurotoxicity and stress-related disorders. Developmental exposures at target concentrations did not significantly alter survival, overall development, or background activity, and had minor effects on developmental morphology. Sodium arsenate and monomethylarsonic acid exaggerated the behavioral responses of larval zebrafish, whereas sodium arsenite depressed them. Sodium arsenate induced significant effects on molecular biomarkers. This study highlights the effects of developmental exposure to arsenicals on the behavioral stress response, the role chemical formulation plays in exerting toxicological effects, and the possible association with stress-related disorders.
Collapse
Affiliation(s)
- Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, United States
| |
Collapse
|
3
|
Fagundes T, Pannetier P, Gölz L, Behnstedt L, Morthorst J, Vergauwen L, Knapen D, Holbech H, Braunbeck T, Baumann L. The generation gap in endocrine disruption: Can the integrated fish endocrine disruptor test (iFEDT) bridge the gap by assessing intergenerational effects of thyroid hormone system disruption? AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 272:106969. [PMID: 38824743 DOI: 10.1016/j.aquatox.2024.106969] [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: 12/12/2023] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/04/2024]
Abstract
Thyroid hormones (THs) act early in ontogenesis, even prior to the differentiation of thyrocytes. Maternal transfer of THs is therefore known to play an essential role in early development. Current OECD test guidelines for the assessment of TH system disruption (THSD) do not address inter- or transgenerational effects. The integrated fish endocrine disruptor test (iFEDT), a test combining parental and developmental exposure of filial fish, may fill this gap. We tested the ability of the iFEDT to detect intergenerational effects in zebrafish (Danio rerio): Parental fish were exposed to propylthiouracil (PTU), an inhibitor of TH synthesis, or not exposed. The offspring was submitted to a crossed experimental design to obtain four exposure scenarios: (1) no exposure at all, (2) parental exposure only, (3) embryonic exposure only, and (4) combined parental and embryonic exposure. Swim bladder inflation, visual motor response (VMR) and gene expression of the progeny were analysed. Parental, but not embryonic PTU exposure reduced the size of the swim bladder of 5 d old embryos, indicating the existence of intergenerational effects. The VMR test produced opposite responses in 4.5 d old embryos exposed to PTU vs. embryos derived from exposed parents. Embryonic exposure, but not parental exposure increased gene expression of thyroperoxidase, the target of PTU, most likely due to a compensatory mechanism. The gene expression of pde-6h (phosphodiesterase) was reduced by embryonic, but not parental exposure, suggesting downregulation of phototransduction pathways. Hence, adverse effects on swim bladder inflation appear more sensitive to parental than embryonic exposure and the iFEDT represents an improvement in the testing strategy for THSD.
Collapse
Affiliation(s)
- Teresa Fagundes
- Aquatic Ecology and Toxicology Group, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany; Eurofins Aquatic Ecotoxicolgy, Eutinger Str. 24, D-75223 Niefern-Öschelbronn, Germany
| | - Pauline Pannetier
- Aquatic Ecology and Toxicology Group, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany; Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail, Laboratoire de Ploufragan-Plouzané-Niort, Site de Plouzané, Technopôle Brest Iroise, CS 10070, F-29280 Plouzané, France
| | - Lisa Gölz
- Aquatic Ecology and Toxicology Group, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany; Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Laura Behnstedt
- Aquatic Ecology and Toxicology Group, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany
| | - Jane Morthorst
- University of Southern Denmark, Institute of Biology, Campusvej 55, DK-5230 Odense M, Denmark
| | - Lucia Vergauwen
- University of Antwerp, Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, Universiteitsplein 1, BE-2160 Wilrijk, Belgium
| | - Dries Knapen
- University of Antwerp, Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, Universiteitsplein 1, BE-2160 Wilrijk, Belgium
| | - Henrik Holbech
- University of Southern Denmark, Institute of Biology, Campusvej 55, DK-5230 Odense M, Denmark
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Group, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany
| | - Lisa Baumann
- Aquatic Ecology and Toxicology Group, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, D-69120 Heidelberg, Germany; Amsterdam Institute for Life and Environment, Section Environmental Health & Toxicology, Vrije Universiteit Amsterdam, De Boelelaan 1085, NL-1081 HV Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Huang J, Sun C, Huang Z, Zhu Y, Chen SX. Upregulation of coagulation factor V by glucocorticoid in the preovulatory follicles of zebrafish. J Steroid Biochem Mol Biol 2024; 241:106521. [PMID: 38631601 PMCID: PMC11140551 DOI: 10.1016/j.jsbmb.2024.106521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/06/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
Increased cortisol levels in the preovulatory follicular fluid suggests a role of glucocorticoid in human ovulation. However, the mechanisms through which cortisol regulates the ovulatory process remain poorly understood. In this study, we examined the upregulation of f5 mRNA by glucocorticoid and its receptor (Gr) in the preovulatory follicles of zebrafish. Our findings demonstrate a significant increase in 11β-hydroxysteroid dehydrogenase type 2 (hsd11b2), a cortisol response gene, in preovulatory follicles. Additionally, hydrocortisone exerts a dose- and time-dependent upregulation of f5 mRNA in these follicles. Importantly, this stimulatory effect is Gr-dependent, as it was completely abolished in gr-/- mutants. Furthermore, site-directed mutagenesis identified a glucocorticoid response element (GRE) in the promoter of zebrafish f5. Interestingly, successive incubation of hydrocortisone and the native ovulation-inducing steroid, progestin (17α,20β-dihydroxy-4-pregnen-3-one, DHP), further enhanced f5 expression in preovulatory follicles. Overall, our results indicate that the dramatic increase of f5 expression in preovulatory follicles is partially attributable to the regulation of glucocorticoid and Gr.
Collapse
Affiliation(s)
- Jing Huang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Chao Sun
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhuo Huang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China; Department of Biology, East Carolina University, 101 E. 10th Street, Greenville, NC 27858, USA
| | - Shi Xi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
5
|
McAtee D, Abdelmoneim A. A zebrafish-based acoustic motor response (AMR) assay to evaluate chemical-induced developmental neurotoxicity. Neurotoxicology 2024; 103:60-70. [PMID: 38851595 DOI: 10.1016/j.neuro.2024.06.003] [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: 08/21/2023] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Behavioral assays using early-developing zebrafish (Danio rerio) offer a valuable supplement to the in vitro battery adopted as new approach methodologies (NAMs) for assessing risk of chemical-induced developmental neurotoxicity. However, the behavioral assays primarily adopted rely on visual stimulation to elicit behavioral responses, known as visual motor response (VMR) assays. Ocular deficits resulting from chemical exposures can, therefore, confound the behavioral responses, independent of effects on the nervous system. This highlights the need for complementary assays employing alternative forms of sensory stimulation. In this study, we investigated the efficacy of acoustic stimuli as triggers of behavioral responses in larval zebrafish, determined the most appropriate data acquisition mode, and evaluated the suitability of an acoustic motor response (AMR) assay as means to assess alterations in brain activity and risk of chemical-induced developmental neurotoxicity. We quantified the motor responses of 120 h post-fertilization (hpf) larvae to acoustic stimuli with varying patterns and frequencies, and determined the optimal time intervals for data acquisition. Following this, we examined changes in acoustic and visual motor responses resulting from exposures to pharmacological agents known to impact brain activity (pentylenetetrazole (PTZ) and tricaine-s (MS-222)). Additionally, we examined the AMR and VMR of larvae following exposure to two environmental contaminants associated with developmental neurotoxicity: arsenic (As) and cadmium (Cd). Our findings indicate that exposure to a 100 Hz sound frequency in 100 ms pulses elicits the strongest behavioral response among the acoustic stimuli tested and data acquisition in 2 s time intervals is suitable for response assessment. Exposure to PTZ exaggerated and depressed both AMR and VMR in a concentration-dependent manner, while exposure to MS-222 only depressed them. Similarly, exposure to As and Cd induced respective hyper- and hypo-activation of both motor responses. This study highlights the efficiency of the proposed zebrafish-based AMR assay in demonstrating risk of chemical-induced developmental neurotoxicity and its suitability as a complement to the widely adopted VMR assay.
Collapse
Affiliation(s)
- Demetrius McAtee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Ahmed Abdelmoneim
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
6
|
Hong Y, Sourander C, Hackl B, Patton JS, John J, Paatero I, Coffey E. Jnk1 and downstream signalling hubs regulate anxiety-like behaviours in a zebrafish larvae phenotypic screen. Sci Rep 2024; 14:11174. [PMID: 38750129 PMCID: PMC11096340 DOI: 10.1038/s41598-024-61337-3] [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: 01/02/2024] [Accepted: 05/04/2024] [Indexed: 05/18/2024] Open
Abstract
Current treatments for anxiety and depression show limited efficacy in many patients, indicating the need for further research into the underlying mechanisms. JNK1 has been shown to regulate anxiety- and depressive-like behaviours in mice, however the effectors downstream of JNK1 are not known. Here we compare the phosphoproteomes from wild-type and Jnk1-/- mouse brains and identify JNK1-regulated signalling hubs. We next employ a zebrafish (Danio rerio) larvae behavioural assay to identify an antidepressant- and anxiolytic-like (AA) phenotype based on 2759 measured stereotypic responses to clinically proven antidepressant and anxiolytic (AA) drugs. Employing machine learning, we classify an AA phenotype from extracted features measured during and after a startle battery in fish exposed to AA drugs. Using this classifier, we demonstrate that structurally independent JNK inhibitors replicate the AA phenotype with high accuracy, consistent with findings in mice. Furthermore, pharmacological targeting of JNK1-regulated signalling hubs identifies AKT, GSK-3, 14-3-3 ζ/ε and PKCε as downstream hubs that phenocopy clinically proven AA drugs. This study identifies AKT and related signalling molecules as mediators of JNK1-regulated antidepressant- and anxiolytic-like behaviours. Moreover, the assay shows promise for early phase screening of compounds with anti-stress-axis properties and for mode of action analysis.
Collapse
Affiliation(s)
- Ye Hong
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Christel Sourander
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Benjamin Hackl
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Jedidiah S Patton
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Jismi John
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland
| | - Eleanor Coffey
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520, Turku, Finland.
| |
Collapse
|
7
|
Pietsch C, Konrad J, Wernicke von Siebenthal E, Pawlak P. Multiple faces of stress in the zebrafish ( Danio rerio) brain. Front Physiol 2024; 15:1373234. [PMID: 38711953 PMCID: PMC11070943 DOI: 10.3389/fphys.2024.1373234] [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: 01/19/2024] [Accepted: 03/19/2024] [Indexed: 05/08/2024] Open
Abstract
The changing expressions of certain genes as a consequence of exposure to stressors has not been studied in detail in the fish brain. Therefore, a stress trial with zebrafish was conducted, aiming at identifying relevant gene regulation pathways in different regions of the brain. As acute stressors within this trial, feed rewarding, feed restriction, and air exposure have been used. The gene expression data from the experimental fish brains have been analyzed by means of principal component analyses (PCAs), whereby the individual genes have been compiled according to the regulation pathways in the brain. The results did not indicate a mutual response across the treatment and gender groups. To evaluate whether a similar sample structure belonging to a large sample size would have allowed the classification of the gene expression patterns according to the treatments, the data have been bootstrapped and used for building random forest models. These revealed a high accuracy of the classifications, but different genes in the female and male zebrafish were found to have contributed to the classification algorithms the most. These analyses showed that less than eight genes are, in most cases, sufficient for an accurate classification. Moreover, mainly genes belonging to the stress axis, to the isotocin regulation pathways, or to the serotonergic pathways had the strongest influence on the outcome of the classification models.
Collapse
Affiliation(s)
- Constanze Pietsch
- School of Agricultural, Forest and Food Sciences (HAFL), University of Applied Sciences Bern (BFH), Zollikofen, Switzerland
| | - Jonathan Konrad
- School of Agricultural, Forest and Food Sciences (HAFL), University of Applied Sciences Bern (BFH), Zollikofen, Switzerland
| | - Elena Wernicke von Siebenthal
- School of Agricultural, Forest and Food Sciences (HAFL), University of Applied Sciences Bern (BFH), Zollikofen, Switzerland
| | - Paulina Pawlak
- School of Agricultural, Forest and Food Sciences (HAFL), University of Applied Sciences Bern (BFH), Zollikofen, Switzerland
- Division of Behavioural Ecology, Institute of Ecology and Evolution, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Eachus H, Choi MK, Tochwin A, Kaspareit J, Ho M, Ryu S. Elevated glucocorticoid alters the developmental dynamics of hypothalamic neurogenesis in zebrafish. Commun Biol 2024; 7:416. [PMID: 38580727 PMCID: PMC10997759 DOI: 10.1038/s42003-024-06060-5] [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: 07/10/2023] [Accepted: 03/16/2024] [Indexed: 04/07/2024] Open
Abstract
Exposure to excess glucocorticoid (GC) during early development is implicated in adult dysfunctions. Reduced adult hippocampal neurogenesis is a well-known consequence of exposure to early life stress or elevated GC, however the effects on neurogenesis during development and effects on other brain regions are not well understood. Using an optogenetic zebrafish model, here we analyse the effects of GC exposure on neurogenesis during development in the whole brain. We identify that the hypothalamus is a highly GC-sensitive region where elevated GC causes precocious development. This is followed by failed maturation and early decline accompanied by impaired feeding, growth, and survival. In GC-exposed animals, the developmental trajectory of hypothalamic progenitor cells is strikingly altered, potentially mediated by direct regulation of transcription factors such as rx3 by GC. Our data provide cellular and molecular level insight into GC-induced alteration of the hypothalamic developmental trajectory, a process crucial for health across the life-course.
Collapse
Affiliation(s)
- Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
- Institute of Health and Neurodevelopment & Aston Pharmacy School, Aston University, Birmingham, B4 7ET, UK
| | - Min-Kyeung Choi
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Anna Tochwin
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK
| | - Johanna Kaspareit
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - May Ho
- Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Soojin Ryu
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
9
|
Lee HB, Shams S, Dang Thi VH, Boyum GE, Modhurima R, Hall EM, Green IK, Cervantes EM, Miguez FE, Clark KJ. Key HPI axis receptors facilitate light adaptive behavior in larval zebrafish. Sci Rep 2024; 14:7759. [PMID: 38565594 PMCID: PMC10987622 DOI: 10.1038/s41598-024-57707-6] [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: 08/06/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
The vertebrate stress response (SR) is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and contributes to generating context appropriate physiological and behavioral changes. Although the HPA axis plays vital roles both in stressful and basal conditions, research has focused on the response under stress. To understand broader roles of the HPA axis in a changing environment, we characterized an adaptive behavior of larval zebrafish during ambient illumination changes. Genetic abrogation of glucocorticoid receptor (nr3c1) decreased basal locomotor activity in light and darkness. Some key HPI axis receptors (mc2r [ACTH receptor], nr3c1), but not nr3c2 (mineralocorticoid receptor), were required to adapt to light more efficiently but became dispensable when longer illumination was provided. Such light adaptation was more efficient in dimmer light. Our findings show that the HPI axis contributes to the SR, facilitating the phasic response and maintaining an adapted basal state, and that certain adaptations occur without HPI axis activity.
Collapse
Affiliation(s)
- Han B Lee
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Soaleha Shams
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Viet Ha Dang Thi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Grace E Boyum
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Rodsy Modhurima
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Emma M Hall
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Izzabella K Green
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Karl J Clark
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.
- Neuroscience, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA.
- Department of Animal Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
10
|
Oglesby Z, Rillorta AN, Agos C, Borges K, Cabradilla S, Garvin M, Higuchi B, Kamaka E, Law C, Liu M, Matsumoto G, Ng T, Quiroz G, Ramiro C, Saito J, Williams M, Yamada A, Yogi Z, Olson S, Shams S, Withy K, Pierret C. Exploring the Hawaiian Ala Wai Watershed with Zebrafish. Zebrafish 2024; 21:206-213. [PMID: 38621213 PMCID: PMC11035842 DOI: 10.1089/zeb.2023.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
The Ala Wai Canal is an artificial waterway in the tourist district of Waikiki in Honolulu, HI. Originally built to collect runoff from industrial, residential, and green spaces dedicated to recreation, the Ala Wai Canal has since experienced potent levels of toxicity due to this runoff entering the watershed and making it hazardous for both marine life and humans at current concentration, including Danio rerio (zebrafish). A community of learners at educations levels from high school to postbaccalaureate from Oahu, HI was connected through the Consortium for Increasing Research and Collaborative Learning Experiences (CIRCLE) distance research program. This team conducted research with an Investigator and team from Mayo Clinic in Rochester, MN, with the Ala Wai Canal as its primary subject. Through CIRCLE, research trainees sent two 32 oz bottles of Ala Wai- acquired water to a partnered laboratory at the Mayo Clinic in which zebrafish embryos were observed at differing concentrations of the sampled water against a variety of developmental and behavioral assays. Research trainees also created atlases of developmental outcomes in zebrafish following exposure to environmental toxins and tables of potential pesticide contaminants to enable the identification of the substances linked to structural defects and enhanced stress during Ala Wai water exposure experiments.
Collapse
Affiliation(s)
- Zachary Oglesby
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
- Hawaii/Pacific Basin Area Health Education Center (AHEC) Department, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, Hawaii, USA
| | - Alanna Nicole Rillorta
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
- Hawaii/Pacific Basin Area Health Education Center (AHEC) Department, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, Hawaii, USA
| | - Cheydon Agos
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Ku'uipo Borges
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Saien Cabradilla
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Michael Garvin
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Bryn Higuchi
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Elisabeth Kamaka
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Chancen Law
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Matthew Liu
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Grace Matsumoto
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Tiffany Ng
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Gemma Quiroz
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Chelsea Ramiro
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Jamie Saito
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Malia Williams
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Asia Yamada
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Zane Yogi
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Sidney Olson
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Soaleha Shams
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kelley Withy
- Consortium For Increasing Learning Research and Collaborative Learning Experience Educational Research Project (CIRCLE Grant), Hawaii/Pacific Basin Area Health Education Center (AHEC), University of Hawai'i at Manoa, Honolulu, Hawaii, USA
- Hawaii/Pacific Basin Area Health Education Center (AHEC) Department, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, Hawaii, USA
| | - Chris Pierret
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Faught E, Schaaf MJM. Molecular mechanisms of the stress-induced regulation of the inflammatory response in fish. Gen Comp Endocrinol 2024; 345:114387. [PMID: 37788784 DOI: 10.1016/j.ygcen.2023.114387] [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: 07/14/2023] [Revised: 09/10/2023] [Accepted: 09/30/2023] [Indexed: 10/05/2023]
Abstract
Stressors in the environment of aquatic organisms can profoundly affect their immune system. The stress response in fish involves the activation of the hypothalamus-pituitary-interrenal (HPI) axis, leading to the release of several stress hormones, among them glucocorticoids, such as cortisol, which bind and activate corticosteroid receptors, namely the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR). These receptors are highly expressed on immune cells, thereby allowing stress to have a potent effect that is classically considered to suppress immune function. In this review, we highlight the conserved structure and function of GR and MR among vertebrates and describe their role in modulating inflammation by regulating the expression of pro-inflammatory and anti-inflammatory genes. In particular, the involvement of MR during inflammation is reviewed, which in many studies has been shown to be immune-enhancing. In recent years, the use of zebrafish as a model organism has opened up new possibilities to study the effects of stress on inflammation, making it possible to investigate knockout lines for MR and/or GR, in combination with transgenic models with fluorescently labeled leukocyte subpopulations that enable the visualization and manipulation of these immune cells. The potential roles of other hormones of the HPI axis, such as corticotrophin-releasing hormone (Crh) and adrenocorticotropic hormone (Acth), in immune modulation are also discussed. Overall, this review highlights the need for further research to elucidate the specific roles of GR, MR and other stress hormones in regulating immune function in fish. Understanding these mechanisms will contribute to improving fish health and advancing our knowledge of stress signalling.
Collapse
Affiliation(s)
- Erin Faught
- Institute of Biology Leiden, Leiden University, The Netherlands
| | | |
Collapse
|
12
|
Lee H, Shams S, Dang Thi VH, Boyum G, Modhurima R, Hall E, Green I, Cervantes E, Miguez F, Clark K. The canonical HPA axis facilitates and maintains light adaptive behavior. RESEARCH SQUARE 2023:rs.3.rs-3240080. [PMID: 37720015 PMCID: PMC10503838 DOI: 10.21203/rs.3.rs-3240080/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The vertebrate stress response (SR) is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and contributes to generating context appropriate physiological and behavioral changes. Although the HPA axis plays vital roles both in stressful and basal conditions, research has focused on the response under stress. To understand broader roles of the HPA axis in a changing environment, we characterized an adaptive behavior of larval zebrafish during ambient illumination changes. The glucocorticoid receptor (nr3c1) was necessary to maintain basal locomotor activity in light and darkness. The HPA axis was required to adapt to light more efficiently but became dispensable when longer illumination was provided. Light adaptation was more efficient in dimmer light and did not require the mineralocorticoid receptor (nr3c2). Our findings show that the HPA axis contributes to the SR at various stages, facilitating the phasic response and maintaining an adapted basal state, and that certain adaptations occur without HPA axis activity.
Collapse
|
13
|
Magierecka A, Cooper B, Sloman KA, Metcalfe NB. Unpredictability of maternal environment shapes offspring behaviour without affecting stress-induced cortisol in an annual vertebrate. Horm Behav 2023; 154:105396. [PMID: 37399780 DOI: 10.1016/j.yhbeh.2023.105396] [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: 10/05/2022] [Revised: 05/23/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
Exposure of females to stressful conditions during pregnancy or oogenesis has a profound effect on the phenotype of their offspring. For example, offspring behavioural phenotype may show altered patterns in terms of the consistency of behavioural patterns and their average level of performance. Maternal stress can also affect the development of the stress axis in offspring leading to alterations in their physiological stress response. However, the majority of evidence comes from studies utilising acute stressors or exogenous glucocorticoids, and little is known about the effect of chronic maternal stress, particularly in the context of stress lasting throughout entire reproductive lifespan. To bridge this knowledge gap, we exposed female sticklebacks to stressful and unpredictable environmental conditions throughout the breeding season. We quantified the activity, sheltering and anxiety-like behaviour of offspring from three successive clutches of these females, and calculated Intra-class Correlation Coefficients for these behaviours in siblings and half-siblings. We also exposed offspring to an acute stressor and measured their peak cortisol levels. An unpredictable maternal environment had no modifying effect on inter-clutch acute stress responsivity, but resulted in diversification of offspring behaviour, indicated by an increased between-individual variability within families. This may represent a bet-hedging strategy, whereby females produce offspring differing in behavioural phenotype, to increase the chance that some of these offspring will be better at coping with the anticipated conditions.
Collapse
Affiliation(s)
- Agnieszka Magierecka
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, UK.
| | - Ben Cooper
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, UK
| | - Katherine A Sloman
- Institute for Biomedical and Environmental Health Research, University of the West of Scotland, Lanarkshire, UK
| | - Neil B Metcalfe
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, UK
| |
Collapse
|
14
|
Torres-Pérez JV, Anagianni S, Mech AM, Havelange W, García-González J, Fraser SE, Vallortigara G, Brennan CH. baz1b loss-of-function in zebrafish produces phenotypic alterations consistent with the domestication syndrome. iScience 2023; 26:105704. [PMID: 36582821 PMCID: PMC9793288 DOI: 10.1016/j.isci.2022.105704] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/15/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
BAZ1B is a ubiquitously expressed nuclear protein with roles in chromatin remodeling, DNA replication and repair, and transcription. Reduced BAZ1B expression disrupts neuronal and neural crest development. Variation in the activity of BAZ1B has been proposed to underly morphological and behavioral aspects of domestication through disruption of neural crest development. Knockdown of baz1b in Xenopus embryos and Baz1b loss-of-function (LoF) in mice leads to craniofacial defects consistent with this hypothesis. We generated baz1b LoF zebrafish using CRISPR/Cas9 gene editing to test the hypothesis that baz1b regulates behavioral phenotypes associated with domestication in addition to craniofacial features. Zebrafish with baz1b LoF show mild underdevelopment at larval stages and distinctive craniofacial features later in life. Mutant zebrafish show reduced anxiety-associated phenotypes and an altered ontogeny of social behaviors. Thus, in zebrafish, developmental deficits in baz1b recapitulate both morphological and behavioral phenotypes associated with the domestication syndrome in other species.
Collapse
Affiliation(s)
- Jose V. Torres-Pérez
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
- Departament de Biologia Cel·lular, Biologia Funcional i Antropologia física, Fac. de CC. Biològiques, Universitat de València, C/ Dr. Moliner 50, Burjassot, València 46100, Spain
| | - Sofia Anagianni
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Aleksandra M. Mech
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - William Havelange
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Judit García-González
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
- Department of Genetics and Genomic Sciences, Icahn School of Medicine, Mount Sinai, New York, NY 10029, USA
| | - Scott E. Fraser
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | | | - Caroline H. Brennan
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| |
Collapse
|
15
|
Scarlett KR, Lovin LM, Steele WB, Kim S, Brooks BW. Identifying Behavioral Response Profiles of Two Common Larval Fish Models to a Salinity Gradient. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2022; 83:180-192. [PMID: 35976388 DOI: 10.1007/s00244-022-00951-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Salinization of aquatic systems is an emerging global issue projected to increase in magnitude, frequency, and duration with climate change and landscape modifications. To consider influences of salinity on locomotor activity of common fish models, we examined behavioral response profiles of two species, zebrafish (Danio rerio) and fathead minnow (Pimephales promelas), across a gradient of sodium chloride. Following each experiment, behavior was recorded with automated tracking software and then behavioral response variables, including locomotor (e.g., distance traveled, number of movements, duration of movements) and photolocomotor changes, were examined at several speed thresholds (bursting, cruising, freezing) to identify potential salinity responses. Zebrafish responses were significantly (p < 0.05) reduced at the highest treatment level (5.78 g/L) for multiple behavioral endpoints during both dark and light phases; however, fathead minnow responses were more variable and not consistently concentration dependent. Future efforts are needed to understand behavioral response profiles in combination with anthropogenic contaminants and natural toxins across the freshwater to marine continuum, considering salinization of inland waters, sea level rise, and transport of anthropogenic contaminants and algal toxins from inland waters to coastal systems.
Collapse
Affiliation(s)
- Kendall R Scarlett
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research, Baylor University, Waco, TX, 76798, USA
| | - Lea M Lovin
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research, Baylor University, Waco, TX, 76798, USA
| | - W Baylor Steele
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research, Baylor University, Waco, TX, 76798, USA
- Institute of Biomedical Studies, Baylor University, Waco, TX, 76798, USA
| | - Sujin Kim
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research, Baylor University, Waco, TX, 76798, USA
| | - Bryan W Brooks
- Department of Environmental Science, Center for Reservoir and Aquatic Systems Research, Baylor University, Waco, TX, 76798, USA.
- Institute of Biomedical Studies, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
16
|
Tan JXM, Ang RJW, Wee CL. Larval Zebrafish as a Model for Mechanistic Discovery in Mental Health. Front Mol Neurosci 2022; 15:900213. [PMID: 35813062 PMCID: PMC9263853 DOI: 10.3389/fnmol.2022.900213] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/25/2022] [Indexed: 12/23/2022] Open
Abstract
Animal models are essential for the discovery of mechanisms and treatments for neuropsychiatric disorders. However, complex mental health disorders such as depression and anxiety are difficult to fully recapitulate in these models. Borrowing from the field of psychiatric genetics, we reiterate the framework of 'endophenotypes' - biological or behavioral markers with cellular, molecular or genetic underpinnings - to reduce complex disorders into measurable behaviors that can be compared across organisms. Zebrafish are popular disease models due to the conserved genetic, physiological and anatomical pathways between zebrafish and humans. Adult zebrafish, which display more sophisticated behaviors and cognition, have long been used to model psychiatric disorders. However, larvae (up to 1 month old) are more numerous and also optically transparent, and hence are particularly suited for high-throughput screening and brain-wide neural circuit imaging. A number of behavioral assays have been developed to quantify neuropsychiatric phenomena in larval zebrafish. Here, we will review these assays and the current knowledge regarding the underlying mechanisms of their behavioral readouts. We will also discuss the existing evidence linking larval zebrafish behavior to specific human behavioral traits and how the endophenotype framework can be applied. Importantly, many of the endophenotypes we review do not solely define a diseased state but could manifest as a spectrum across the general population. As such, we make the case for larval zebrafish as a promising model for extending our understanding of population mental health, and for identifying novel therapeutics and interventions with broad impact.
Collapse
Affiliation(s)
| | | | - Caroline Lei Wee
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
17
|
Lee CJ, Paull GC, Tyler CR. Improving zebrafish laboratory welfare and scientific research through understanding their natural history. Biol Rev Camb Philos Soc 2022; 97:1038-1056. [PMID: 34983085 PMCID: PMC9303617 DOI: 10.1111/brv.12831] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
Globally, millions of zebrafish (Danio rerio) are used for scientific laboratory experiments for which researchers have a duty of care, with legal obligations to consider their welfare. Considering the growing use of the zebrafish as a vertebrate model for addressing a diverse range of scientific questions, optimising their laboratory conditions is of major importance for both welfare and improving scientific research. However, most guidelines for the care and breeding of zebrafish for research are concerned primarily with maximising production and minimising costs and pay little attention to the effects on welfare of the environments in which the fish are maintained, or how those conditions affect their scientific research. Here we review the physical and social conditions in which laboratory zebrafish are kept, identifying and drawing attention to factors likely to affect their welfare and experimental science. We also identify a fundamental lack knowledge of how zebrafish interact with many biotic and abiotic features in their natural environment to support ways to optimise zebrafish health and well-being in the laboratory, and in turn the quality of scientific data produced. We advocate that the conditions under which zebrafish are maintained need to become a more integral part of research and that we understand more fully how they influence experimental outcome and in turn interpretations of the data generated.
Collapse
Affiliation(s)
- Carole J. Lee
- Biosciences, Geoffrey Pope BuildingUniversity of ExeterStocker RoadExeterEX4 4QDU.K.
| | - Gregory C. Paull
- Biosciences, Geoffrey Pope BuildingUniversity of ExeterStocker RoadExeterEX4 4QDU.K.
| | - Charles R. Tyler
- Biosciences, Geoffrey Pope BuildingUniversity of ExeterStocker RoadExeterEX4 4QDU.K.
| |
Collapse
|
18
|
Banerjee P, Saha I, Sarkar D, Maiti AK. Contributions and Limitations of Mitochondria-Targeted and Non-Targeted Antioxidants in the Treatment of Parkinsonism: an Updated Review. Neurotox Res 2022; 40:847-873. [PMID: 35386026 DOI: 10.1007/s12640-022-00501-x] [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: 07/24/2021] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022]
Abstract
As conventional therapeutics can only treat the symptoms of Parkinson's disease (PD), major focus of research in recent times is to slow down or prevent the progression of neuronal degeneration in PD. Non-targeted antioxidants have been an integral part of the conventional therapeutics regimen; however, their importance have lessened over time because of their controversial outcomes in clinical PD trials. Inability to permeate and localize within the mitochondria remains the main drawback on the part of non-targeted antioxidants inspite of possessing free radical scavenging properties. In contrast, mitochondrial-targeted antioxidants (MTAs), a special class of compounds have emerged having high advantages over non-targeted antioxidants by virtue of efficient pharmacokinetics and better absorption rate with capability to localize many fold inside the mitochondrial matrix. Preclinical experimentations indicate that MTAs have the potential to act as better alternatives compared to conventional non-targeted antioxidants in treating PD; however, sufficient clinical trials have not been conducted to investigate the efficacies of MTAs in treating PD. Controversial clinical outcomes on the part of non-targeted antioxidants and lack of clinical trials involving MTAs have made it difficult to go ahead with a direct comparison and in turn have slowed down the progress of development of safer and better alternate strategies in treating PD. This review provides an insight on the roles MTAs and non-targeted antioxidants have played in the treatment of PD till date in preclinical and clinical settings and discusses about the limitations of mitochondria-targeted and non-targeted antioxidants that can be resolved for developing effective strategies in treating Parkinsonism.
Collapse
Affiliation(s)
- Priyajit Banerjee
- Department of Zoology, University of Burdwan, Burdwan, West Bengal, Pin-713104, India
| | - Ishita Saha
- Department of Physiology, Medical College Kolkata, Kolkata, West Bengal, Pin-700073, India
| | - Diptendu Sarkar
- Department of Microbiology, Ramakrishna Mission Vidyamandira, Belur Math, Howrah, West Bengal, 711202, India
| | - Arpan Kumar Maiti
- Mitochondrial Biology and Experimental Therapeutics Laboratory, Department of Zoology, University of North Bengal, District - Darjeeling, P.O. N.B.U, Raja Rammohunpur, West Bengal, Pin-734013, India.
| |
Collapse
|
19
|
Simone BW, Lee HB, Daby CL, Ata H, Restrepo-Castillo S, Martínez-Gálvez G, Kar B, Gendron WA, Clark KJ, Ekker SC. Chimeric RNA: DNA TracrRNA Improves Homology-Directed Repair In Vitro and In Vivo. CRISPR J 2022; 5:40-52. [PMID: 34935462 PMCID: PMC8892967 DOI: 10.1089/crispr.2021.0087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Nearly 90% of human pathogenic mutations are caused by small genetic variations, and methods to correct these errors efficiently are critically important. One way to make small DNA changes is providing a single-stranded oligo deoxynucleotide (ssODN) containing an alteration coupled with a targeted double-strand break (DSB) at the target locus in the genome. Coupling an ssODN donor with a CRISPR-Cas9-mediated DSB is one of the most streamlined approaches to introduce small changes. However, in many systems, this approach is inefficient and introduces imprecise repair at the genetic junctions. We herein report a technology that uses spatiotemporal localization of an ssODN with CRISPR-Cas9 to improve gene alteration. We show that by fusing an ssODN template to the trans-activating RNA (tracrRNA), we recover precise genetic alterations, with increased integration and precision in vitro and in vivo. Finally, we show that this technology can be used to enhance gene conversion with other gene editing tools such as transcription activator like effector nucleases.
Collapse
Affiliation(s)
- Brandon W. Simone
- Department of Biochemistry and Molecular Biology, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Han B. Lee
- Department of Biochemistry and Molecular Biology, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Camden L. Daby
- Department of Biochemistry and Molecular Biology, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Hirotaka Ata
- Department of Clinical and Translational Sciences, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Santiago Restrepo-Castillo
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Track, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Gabriel Martínez-Gálvez
- Mayo Clinic Graduate School of Biomedical Sciences, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Bibekananda Kar
- Department of Biochemistry and Molecular Biology, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - William A.C. Gendron
- Mayo Clinic Graduate School of Biomedical Sciences, Virology and Gene Therapy Track, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Karl J. Clark
- Department of Biochemistry and Molecular Biology, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Biomedical Engineering and Physiology Track, Mayo Clinic, Rochester, Minnesota, USA
- Address correspondence to: Stephen C. Ekker, PhD, Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA,
| |
Collapse
|
20
|
Mech AM, Merteroglu M, Sealy IM, Teh MT, White RJ, Havelange W, Brennan CH, Busch-Nentwich EM. Behavioral and Gene Regulatory Responses to Developmental Drug Exposures in Zebrafish. Front Psychiatry 2022; 12:795175. [PMID: 35082702 PMCID: PMC8785235 DOI: 10.3389/fpsyt.2021.795175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/29/2021] [Indexed: 01/22/2023] Open
Abstract
Developmental consequences of prenatal drug exposure have been reported in many human cohorts and animal studies. The long-lasting impact on the offspring-including motor and cognitive impairments, cranial and cardiac anomalies and increased prevalence of ADHD-is a socioeconomic burden worldwide. Identifying the molecular changes leading to developmental consequences could help ameliorate the deficits and limit the impact. In this study, we have used zebrafish, a well-established behavioral and genetic model with conserved drug response and reward pathways, to identify changes in behavior and cellular pathways in response to developmental exposure to amphetamine, nicotine or oxycodone. In the presence of the drug, exposed animals showed altered behavior, consistent with effects seen in mammalian systems, including impaired locomotion and altered habituation to acoustic startle. Differences in responses seen following acute and chronic exposure suggest adaptation to the presence of the drug. Transcriptomic analysis of exposed larvae revealed differential expression of numerous genes and alterations in many pathways, including those related to cell death, immunity and circadian rhythm regulation. Differential expression of circadian rhythm genes did not correlate with behavioral changes in the larvae, however, two of the circadian genes, arntl2 and per2, were also differentially expressed at later stages of development, suggesting a long-lasting impact of developmental exposures on circadian gene expression. The immediate-early genes, egr1, egr4, fosab, and junbb, which are associated with synaptic plasticity, were downregulated by all three drugs and in situ hybridization showed that the expression for all four genes was reduced across all neuroanatomical regions, including brain regions implicated in reward processing, addiction and other psychiatric conditions. We anticipate that these early changes in gene expression in response to drug exposure are likely to contribute to the consequences of prenatal exposure and their discovery might pave the way to therapeutic intervention to ameliorate the long-lasting deficits.
Collapse
Affiliation(s)
- Aleksandra M. Mech
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
| | - Munise Merteroglu
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Ian M. Sealy
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - Muy-Teck Teh
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, England, United Kingdom
| | - Richard J. White
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| | - William Havelange
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
| | - Caroline H. Brennan
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
| | - Elisabeth M. Busch-Nentwich
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London, United Kingdom
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Lu Y, Shi C, Jin X, He J, Yin Z. Domestication of farmed fish via the attenuation of stress responses mediated by the hypothalamus-pituitary-inter-renal endocrine axis. Front Endocrinol (Lausanne) 2022; 13:923475. [PMID: 35937837 PMCID: PMC9353172 DOI: 10.3389/fendo.2022.923475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022] Open
Abstract
Human-directed domestication of terrestrial animals traditionally requires thousands of years for breeding. The most prominent behavioral features of domesticated animals include reduced aggression and enhanced tameness relative to their wild forebears, and such behaviors improve the social tolerance of domestic animals toward both humans and crowds of their own species. These behavioral responses are primarily mediated by the hypothalamic-pituitary-adrenal (inter-renal in fish) (HPA/I) endocrine axis, which is involved in the rapid conversion of neuronal-derived perceptual information into hormonal signals. Over recent decades, growing evidence implicating the attenuation of the HPA/I axis during the domestication of animals have been identified through comprehensive genomic analyses of the paleogenomic datasets of wild progenitors and their domestic congeners. Compared with that of terrestrial animals, domestication of most farmed fish species remains at early stages. The present review focuses on the application of HPI signaling attenuation to accelerate the domestication and genetic breeding of farmed fish. We anticipate that deeper understanding of HPI signaling and its implementation in the domestication of farmed fish will benefit genetic breeding to meet the global demands of the aquaculture industry.
Collapse
Affiliation(s)
- Yao Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Chuang Shi
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Xia Jin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jiangyan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhan Yin
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
- Hubei Hongshan Laboratory, Wuhan, China
- Hainan Yazhou Bay Seed Laboratory, Sanya, China
- *Correspondence: Zhan Yin,
| |
Collapse
|
22
|
Gans IM, Coffman JA. Glucocorticoid-Mediated Developmental Programming of Vertebrate Stress Responsivity. Front Physiol 2021; 12:812195. [PMID: 34992551 PMCID: PMC8724051 DOI: 10.3389/fphys.2021.812195] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023] Open
Abstract
Glucocorticoids, vertebrate steroid hormones produced by cells of the adrenal cortex or interrenal tissue, function dynamically to maintain homeostasis under constantly changing and occasionally stressful environmental conditions. They do so by binding and thereby activating nuclear receptor transcription factors, the Glucocorticoid and Mineralocorticoid Receptors (MR and GR, respectively). The GR, by virtue of its lower affinity for endogenous glucocorticoids (cortisol or corticosterone), is primarily responsible for transducing the dynamic signals conveyed by circadian and ultradian glucocorticoid oscillations as well as transient pulses produced in response to acute stress. These dynamics are important determinants of stress responsivity, and at the systemic level are produced by feedforward and feedback signaling along the hypothalamus-pituitary-adrenal/interrenal axis. Within receiving cells, GR signaling dynamics are controlled by the GR target gene and negative feedback regulator fkpb5. Chronic stress can alter signaling dynamics via imperfect physiological adaptation that changes systemic and/or cellular set points, resulting in chronically elevated cortisol levels and increased allostatic load, which undermines health and promotes development of disease. When this occurs during early development it can "program" the responsivity of the stress system, with persistent effects on allostatic load and disease susceptibility. An important question concerns the glucocorticoid-responsive gene regulatory network that contributes to such programming. Recent studies show that klf9, a ubiquitously expressed GR target gene that encodes a Krüppel-like transcription factor important for metabolic plasticity and neuronal differentiation, is a feedforward regulator of GR signaling impacting cellular glucocorticoid responsivity, suggesting that it may be a critical node in that regulatory network.
Collapse
Affiliation(s)
- Ian M. Gans
- MDI Biological Laboratory, Salisbury Cove, ME, United States
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - James A. Coffman
- MDI Biological Laboratory, Salisbury Cove, ME, United States
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| |
Collapse
|
23
|
Homeostatic Regulation of Glucocorticoid Receptor Activity by Hypoxia-Inducible Factor 1: From Physiology to Clinic. Cells 2021; 10:cells10123441. [PMID: 34943949 PMCID: PMC8699886 DOI: 10.3390/cells10123441] [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: 11/12/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs) represent a well-known class of lipophilic steroid hormones biosynthesised, with a circadian rhythm, by the adrenal glands in humans and by the inter-renal tissue in teleost fish (e.g., zebrafish). GCs play a key role in the regulation of numerous physiological processes, including inflammation, glucose, lipid, protein metabolism and stress response. This is achieved through binding to their cognate receptor, GR, which functions as a ligand-activated transcription factor. Due to their potent anti-inflammatory and immune-suppressive action, synthetic GCs are broadly used for treating pathological disorders that are very often linked to hypoxia (e.g., rheumatoid arthritis, inflammatory, allergic, infectious, and autoimmune diseases, among others) as well as to prevent graft rejections and against immune system malignancies. However, due to the presence of adverse effects and GC resistance their therapeutic benefits are limited in patients chronically treated with steroids. For this reason, understanding how to fine-tune GR activity is crucial in the search for novel therapeutic strategies aimed at reducing GC-related side effects and effectively restoring homeostasis. Recent research has uncovered novel mechanisms that inhibit GR function, thereby causing glucocorticoid resistance, and has produced some surprising new findings. In this review we analyse these mechanisms and focus on the crosstalk between GR and HIF signalling. Indeed, its comprehension may provide new routes to develop novel therapeutic targets for effectively treating immune and inflammatory response and to simultaneously facilitate the development of innovative GCs with a better benefits-risk ratio.
Collapse
|
24
|
Tang C, Zhu Y, Laziyan Y, Yang C, He C, Zuo Z. Long-term exposure to cyprodinil causes abnormal zebrafish aggressive and antipredator behavior through the hypothalamic-pituitary-interrenal axis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 241:106002. [PMID: 34717145 DOI: 10.1016/j.aquatox.2021.106002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 06/13/2023]
Abstract
Cyprodinil, one of the main pyrimidinamine fungicides, has been used to control fungal diseases in plants and vegetables worldwide. Previous studies have investigated the influences of cyprodinil on the developmental and reproductive toxicity of fish. However, it remains unknown whether it affects fish behaviors and the underlying mechanisms. In our current study, zebrafish, an ideal model animal for behavioral studies, were exposed to cyprodinil from fertilization to 240 days postfertilization at 0.1 μg/L (environmentally relevant concentration) and 1, 10 μg/L. Firstly, we observed that aggressive behavior of zebrafish was significantly enhanced after exposure to 0.1-10 μg/L cyprodinil and antipredator behavior was decreased after exposure. Cyprodinil exposure altered the adrenocorticotropic hormone and cortisol levels, which regulate cortisol homeostasis and were significantly reduced in all exposure groups (0.1-10 μg/L). In addition, most of the key genes in the hypothalamic-pituitary-interrenal gland axis, such as corticotropin-releasing hormone and melanocortin 2 receptor, were downregulated significantly in all exposure groups, which was consistent with the hormone levels. In addition, in the hypothalamus, the number of apoptotic cells increased in a dose-dependent manner in the cyprodinil exposure groups. Moreover, these changes were potentially responsible for the increased aggression of zebrafish during the mirror-like aggressive test and for the reduced antipredator behavior during the predator avoidance test. Overall, the data provided herein further our understanding of cyprodinil toxicity and can be used to assess the ecological effects of cyprodinil on the induction of abnormal behaviors at the environmental level.
Collapse
Affiliation(s)
- Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Yue Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Yibimu Laziyan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
25
|
Chronic exposure to stressors has a persistent effect on feeding behaviour but not cortisol levels in sticklebacks. Anim Behav 2021. [DOI: 10.1016/j.anbehav.2021.08.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Eachus H, Choi MK, Ryu S. The Effects of Early Life Stress on the Brain and Behaviour: Insights From Zebrafish Models. Front Cell Dev Biol 2021; 9:657591. [PMID: 34368117 PMCID: PMC8335398 DOI: 10.3389/fcell.2021.657591] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/20/2021] [Indexed: 01/27/2023] Open
Abstract
The early life period represents a window of increased vulnerability to stress, during which exposure can lead to long-lasting effects on brain structure and function. This stress-induced developmental programming may contribute to the behavioural changes observed in mental illness. In recent decades, rodent studies have significantly advanced our understanding of how early life stress (ELS) affects brain development and behaviour. These studies reveal that ELS has long-term consequences on the brain such as impairment of adult hippocampal neurogenesis, altering learning and memory. Despite such advances, several key questions remain inadequately answered, including a comprehensive overview of brain regions and molecular pathways that are altered by ELS and how ELS-induced molecular changes ultimately lead to behavioural changes in adulthood. The zebrafish represents a novel ELS model, with the potential to contribute to answering some of these questions. The zebrafish offers some important advantages such as the ability to non-invasively modulate stress hormone levels in a whole animal and to visualise whole brain activity in freely behaving animals. This review discusses the current status of the zebrafish ELS field and its potential as a new ELS model.
Collapse
Affiliation(s)
- Helen Eachus
- Living Systems Institute and College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Min-Kyeung Choi
- Living Systems Institute and College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Soojin Ryu
- Living Systems Institute and College of Medicine and Health, University of Exeter, Exeter, United Kingdom.,Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
27
|
García-González J, de Quadros B, Havelange W, Brock AJ, Brennan CH. Behavioral Effects of Developmental Exposure to JWH-018 in Wild-Type and Disrupted in Schizophrenia 1 ( disc1) Mutant Zebrafish. Biomolecules 2021; 11:biom11020319. [PMID: 33669793 PMCID: PMC7922669 DOI: 10.3390/biom11020319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 11/17/2022] Open
Abstract
Synthetic cannabinoids can cause acute adverse psychological effects, but the potential impact when exposure happens before birth is unknown. Use of synthetic cannabinoids during pregnancy may affect fetal brain development, and such effects could be moderated by the genetic makeup of an individual. Disrupted in schizophrenia 1 (DISC1) is a gene with important roles in neurodevelopment that has been associated with psychiatric disorders in pedigree analyses. Using zebrafish as a model, we investigated (1) the behavioral impact of developmental exposure to 3 μM 1-pentyl-3-(1-naphthoyl)-indole (JWH-018; a common psychoactive synthetic cannabinoid) and (2) whether disc1 moderates the effects of JWH-018. As altered anxiety responses are seen in several psychiatric disorders, we focused on zebrafish anxiety-like behavior. Zebrafish embryos were exposed to JWH-018 from one to six days post-fertilization. Anxiety-like behavior was assessed using forced light/dark and acoustic startle assays in larvae and novel tank diving in adults. Compared to controls, both acutely and developmentally exposed zebrafish larvae had impaired locomotion during the forced light/dark test, but anxiety levels and response to startle stimuli were unaltered. Adult zebrafish developmentally exposed to JWH-018 spent less time on the bottom of the tank, suggesting decreased anxiety. Loss-of-function in disc1 increased anxiety-like behavior in the tank diving assay but did not alter sensitivity to JWH-018. Results suggest developmental exposure to JWH-018 has a long-term behavioral impact in zebrafish, which is not moderated by disc1.
Collapse
Affiliation(s)
- Judit García-González
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK; (J.G.-G.); (B.d.Q.); (W.H.)
| | - Bruno de Quadros
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK; (J.G.-G.); (B.d.Q.); (W.H.)
| | - William Havelange
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK; (J.G.-G.); (B.d.Q.); (W.H.)
| | | | - Caroline H. Brennan
- School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK; (J.G.-G.); (B.d.Q.); (W.H.)
- Correspondence:
| |
Collapse
|
28
|
Zebrafish as a Successful Animal Model for Screening Toxicity of Medicinal Plants. PLANTS 2020; 9:plants9101345. [PMID: 33053800 PMCID: PMC7601530 DOI: 10.3390/plants9101345] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022]
Abstract
The zebrafish (Danio rerio) is used as an embryonic and larval model to perform in vitro experiments and developmental toxicity studies. Zebrafish may be used to determine the toxicity of samples in early screening assays, often in a high-throughput manner. The zebrafish embryotoxicity model is at the leading edge of toxicology research due to the short time required for analyses, transparency of embryos, short life cycle, high fertility, and genetic data similarity. Zebrafish toxicity studies range from assessing the toxicity of bioactive compounds or crude extracts from plants to determining the optimal process. Most of the studied extracts were polar, such as ethanol, methanol, and aqueous solutions, which were used to detect the toxicity and bioactivity. This review examines the latest research using zebrafish as a study model and highlights its power as a tool for detecting toxicity of medicinal plants and its effectiveness at enhancing the understanding of new drug generation. The goal of this review was to develop a link to ethnopharmacological zebrafish studies that can be used by other researchers to conduct future research.
Collapse
|
29
|
A vertebrate model to reveal neural substrates underlying the transitions between conscious and unconscious states. Sci Rep 2020; 10:15789. [PMID: 32978423 PMCID: PMC7519646 DOI: 10.1038/s41598-020-72669-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/28/2020] [Indexed: 12/16/2022] Open
Abstract
The field of neuropharmacology has not yet achieved a full understanding of how the brain transitions between states of consciousness and drug-induced unconsciousness, or anesthesia. Many small molecules are used to alter human consciousness, but the repertoire of underlying molecular targets, and thereby the genes, are incompletely understood. Here we describe a robust larval zebrafish model of anesthetic action, from sedation to general anesthesia. We use loss of movement under three different conditions, spontaneous movement, electrical stimulation or a tap, as a surrogate for sedation and general anesthesia, respectively. Using these behavioral patterns, we find that larval zebrafish respond to inhalational and IV anesthetics at concentrations similar to mammals. Additionally, known sedative drugs cause loss of spontaneous larval movement but not to the tap response. This robust, highly tractable vertebrate model can be used in the detection of genes and neural substrates involved in the transition from consciousness to unconsciousness.
Collapse
|
30
|
Introducing the Amphibious Mudskipper Goby as a Unique Model to Evaluate Neuro/Endocrine Regulation of Behaviors Mediated by Buccal Sensation and Corticosteroids. Int J Mol Sci 2020; 21:ijms21186748. [PMID: 32938015 PMCID: PMC7555618 DOI: 10.3390/ijms21186748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 11/19/2022] Open
Abstract
Some fish have acquired the ability to breathe air, but these fish can no longer flush their gills effectively when out of water. Hence, they have developed characteristic means for defense against external stressors, including thirst (osmolarity/ions) and toxicity. Amphibious fish, extant air-breathing fish emerged from water, may serve as models to examine physiological responses to these stressors. Some of these fish, including mudskipper gobies such as Periophthalmodon schlosseri, Boleophthalmus boddarti and our Periophthalmus modestus, display distinct adaptational behaviors to these factors compared with fully aquatic fish. In this review, we introduce the mudskipper goby as a unique model to study the behaviors and the neuro/endocrine mechanisms of behavioral responses to the stressors. Our studies have shown that a local sensation of thirst in the buccal cavity—this being induced by dipsogenic hormones—motivates these fish to move to water through a forebrain response. The corticosteroid system, which is responsive to various stressors, also stimulates migration, possibly via the receptors in the brain. We suggest that such fish are an important model to deepen insights into the stress-related neuro/endocrine-behavioral effects.
Collapse
|
31
|
Klf9 is a key feedforward regulator of the transcriptomic response to glucocorticoid receptor activity. Sci Rep 2020; 10:11415. [PMID: 32651405 PMCID: PMC7351738 DOI: 10.1038/s41598-020-68040-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/17/2020] [Indexed: 01/01/2023] Open
Abstract
The zebrafish has recently emerged as a model system for investigating the developmental roles of glucocorticoid signaling and the mechanisms underlying glucocorticoid-induced developmental programming. To assess the role of the Glucocorticoid Receptor (GR) in such programming, we used CRISPR-Cas9 to produce a new frameshift mutation, GR369-, which eliminates all potential in-frame initiation codons upstream of the DNA binding domain. Using RNA-seq to ask how this mutation affects the larval transcriptome under both normal conditions and with chronic cortisol treatment, we find that GR mediates most of the effects of the treatment, and paradoxically, that the transcriptome of cortisol-treated larvae is more like that of larvae lacking a GR than that of larvae with a GR, suggesting that the cortisol-treated larvae develop GR resistance. The one transcriptional regulator that was both underexpressed in GR369- larvae and consistently overexpressed in cortisol-treated larvae was klf9. We therefore used CRISPR-Cas9-mediated mutation of klf9 and RNA-seq to assess Klf9-dependent gene expression in both normal and cortisol-treated larvae. Our results indicate that Klf9 contributes significantly to the transcriptomic response to chronic cortisol exposure, mediating the upregulation of proinflammatory genes that we reported previously.
Collapse
|
32
|
Wei P, Zhao F, Zhang X, Ru S. Long-term exposure of zebrafish to bisphenol S impairs stress function of hypothalamic-pituitary-interrenal axis and causes anxiety-like behavioral responses to novelty. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 716:137092. [PMID: 32044495 DOI: 10.1016/j.scitotenv.2020.137092] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/21/2020] [Accepted: 02/01/2020] [Indexed: 06/10/2023]
Abstract
Bisphenol S (BPS), a main substitute of bisphenol A, has been reported to induce multiple endocrine disrupting effects on animals, however, whether it can interfere with the corticosteroid-endocrine system still remains unknown. Furthermore, previous studies mainly investigated the influences of environmental pollutants on corticosteroid levels and gene expressions of hypothalamic-pituitary-interrenal/adrenal (HPI/A) axis, while the downstream toxic effects caused thereafter have not yet been fully elucidated. Considering the key role of cortisol, a primary corticosteroid hormone in teleost, in mediating stress adaptation and the highly positive correlation between cortisol level and anxious phenotype in the novel environment, we hypothesized that an imbalanced cortisol homeostasis due to environmental pollutant exposure may further affect the behavioral responses to novelty stress. In the present study, zebrafish, a valuable model in studying human stress physiology and anxiety behavior, were exposed to BPS from embryos to adults (120 days) at environmentally relevant concentrations (1 and 10 μg/L) and 100 μg/L. Results found that long-term exposure to BPS increased whole-body cortisol levels and caused abnormal expressions of HPI axis genes. Moreover, the excessive cortisol levels may be due to the inhibition of cortisol catabolism and excretion, as evidenced by the down-regulated expressions of hydroxysteroid 11-beta dehydrogenase 2 and hydroxysteroid 20-beta dehydrogenase 2 genes. More importantly, as we speculated, excessive cortisol levels may be responsible for the occurrence of anxiety-like behavioral responses indicated by longer latency, fewer time spent in the upper half, and more erratic movements in a 6-min novel tank test. Overall, our study provides basic data for the comprehensive understanding of BPS toxicity, and emphasizes environmental health risks of BPS in inducing anxiety syndrome at environmentally realistic concentrations.
Collapse
Affiliation(s)
- Penghao Wei
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong province, China
| | - Fei Zhao
- School of Environmental and Municipal Engineering, Qingdao University of Technology, 11 Fushun Road, Qingdao 266033, Shandong province, China
| | - Xiaona Zhang
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong province, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, Shandong province, China.
| |
Collapse
|
33
|
Marchi D, Santhakumar K, Markham E, Li N, Storbeck KH, Krone N, Cunliffe VT, van Eeden FJM. Bidirectional crosstalk between Hypoxia-Inducible Factor and glucocorticoid signalling in zebrafish larvae. PLoS Genet 2020; 16:e1008757. [PMID: 32379754 PMCID: PMC7237044 DOI: 10.1371/journal.pgen.1008757] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/19/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
In the last decades in vitro studies highlighted the potential for crosstalk between Hypoxia-Inducible Factor-(HIF) and glucocorticoid-(GC) signalling pathways. However, how this interplay precisely occurs in vivo is still debated. Here, we use zebrafish larvae (Danio rerio) to elucidate how and to what degree hypoxic signalling affects the endogenous glucocorticoid pathway and vice versa, in vivo. Firstly, our results demonstrate that in the presence of upregulated HIF signalling, both glucocorticoid receptor (Gr) responsiveness and endogenous cortisol levels are repressed in 5 days post fertilisation larvae. In addition, despite HIF activity being low at normoxia, our data show that it already impedes both glucocorticoid activity and levels. Secondly, we further analysed the in vivo contribution of glucocorticoids to HIF activity. Interestingly, our results show that both glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) play a key role in enhancing it. Finally, we found indications that glucocorticoids promote HIF signalling via multiple routes. Cumulatively, our findings allowed us to suggest a model for how this crosstalk occurs in vivo.
Collapse
Affiliation(s)
- Davide Marchi
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
- * E-mail: (DM); (FJMv)
| | - Kirankumar Santhakumar
- Department of Genetic Engineering, SRM Institute of Science and Technology Kattankulathur, India
| | - Eleanor Markham
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Nan Li
- The Bateson Centre & Department of Oncology and Metabolism, School of Medicine, University of Sheffield, Sheffield, United Kingdom
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, Matieland, South Africa
| | - Nils Krone
- The Bateson Centre & Department of Oncology and Metabolism, School of Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Vincent T. Cunliffe
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Fredericus J. M. van Eeden
- The Bateson Centre & Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, United Kingdom
- * E-mail: (DM); (FJMv)
| |
Collapse
|
34
|
Eid JI, Das B. Molecular insights and cell cycle assessment upon exposure to Chaga (Inonotus obliquus) mushroom polysaccharides in zebrafish (Danio rerio). Sci Rep 2020; 10:7406. [PMID: 32366825 PMCID: PMC7198532 DOI: 10.1038/s41598-020-64157-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/06/2020] [Indexed: 02/08/2023] Open
Abstract
Chaga (Inonotus obliquus) mushroom is considered as one of the most powerful antioxidants across the world. Though the therapeutic effects of Chaga components are well characterized in vitro, the in vivo developmental effects are not elucidated in detail. In this study, we assessed the in vivo developmental effects of Chaga polysaccharides in zebrafish, along with revealing the effects on cell cycle and apoptosis. Chaga mushroom polysaccharides comprised xylulose, rhamnose, mannose, glucose, inositol, and galactose, in addition to phenolic compounds; zebrafish embryos exhibited normal embryonic development upon transient exposure to Chaga extract (24 hours). Most embryos (>90%) were found to be healthy even at high concentrations (5 mg/mL). In addition, staining with the DNA binding dye, acridine orange showed that Chaga polysaccharides alleviated oxidative stress. Flow cytometric analysis using H2DCFDA that specifically binds to cells with fragmented DNA showed significantly reduced levels of intracellular reactive oxygen species (ROS) (p < 0.05), which in turn reduced apoptosis in the developing embryos. Cell cycle analysis by measuring the DNA content using flow cytometry revealed that Chaga polysaccharides moderately arrested the cells at G1 stage, thereby inhibiting cell proliferation that can be further explored in cancer studies. Overall, transient exposure of Chaga polysaccharide extract reduced intracellular ROS and assisted in the normal development of zebrafish.
Collapse
Affiliation(s)
- Jehane Ibrahim Eid
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt.
| | - Biswadeep Das
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| |
Collapse
|
35
|
Yilmaz B, Terekeci H, Sandal S, Kelestimur F. Endocrine disrupting chemicals: exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev Endocr Metab Disord 2020; 21:127-147. [PMID: 31792807 DOI: 10.1007/s11154-019-09521-z] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endocrine Disrupting Chemicals (EDCs) are a global problem for environmental and human health. They are defined as "an exogenous chemical, or mixture of chemicals, that can interfere with any aspect of hormone action". It is estimated that there are about 1000 chemicals with endocrine-acting properties. EDCs comprise pesticides, fungicides, industrial chemicals, plasticizers, nonylphenols, metals, pharmaceutical agents and phytoestrogens. Human exposure to EDCs mainly occurs by ingestion and to some extent by inhalation and dermal uptake. Most EDCs are lipophilic and bioaccumulate in the adipose tissue, thus they have a very long half-life in the body. It is difficult to assess the full impact of human exposure to EDCs because adverse effects develop latently and manifest at later ages, and in some people do not present. Timing of exposure is of importance. Developing fetus and neonates are the most vulnerable to endocrine disruption. EDCs may interfere with synthesis, action and metabolism of sex steroid hormones that in turn cause developmental and fertility problems, infertility and hormone-sensitive cancers in women and men. Some EDCs exert obesogenic effects that result in disturbance in energy homeostasis. Interference with hypothalamo-pituitary-thyroid and adrenal axes has also been reported. In this review, potential EDCs, their effects and mechanisms of action, epidemiological studies to analyze their effects on human health, bio-detection and chemical identification methods, difficulties in extrapolating experimental findings and studying endocrine disruptors in humans and recommendations for endocrinologists, individuals and policy makers will be discussed in view of the relevant literature.
Collapse
Affiliation(s)
- Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Hakan Terekeci
- Department of Internal Medicine, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Suleyman Sandal
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Fahrettin Kelestimur
- Department of Endocrinology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
36
|
Lee HB, Schwab TL, Sigafoos AN, Gauerke JL, Krug RG, Serres MR, Jacobs DC, Cotter RP, Das B, Petersen MO, Daby CL, Urban RM, Berry BC, Clark KJ. Novel zebrafish behavioral assay to identify modifiers of the rapid, nongenomic stress response. GENES, BRAIN, AND BEHAVIOR 2019; 18:e12549. [PMID: 30588759 PMCID: PMC6446827 DOI: 10.1111/gbb.12549] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/30/2018] [Accepted: 12/18/2018] [Indexed: 12/23/2022]
Abstract
When vertebrates face acute stressors, their bodies rapidly undergo a repertoire of physiological and behavioral adaptations, which is termed the stress response. Rapid changes in heart rate and blood glucose levels occur via the interaction of glucocorticoids and their cognate receptors following hypothalamic-pituitary-adrenal axis activation. These physiological changes are observed within minutes of encountering a stressor and the rapid time domain rules out genomic responses that require gene expression changes. Although behavioral changes corresponding to physiological changes are commonly observed, it is not clearly understood to what extent hypothalamic-pituitary-adrenal axis activation dictates adaptive behavior. We hypothesized that rapid locomotor response to acute stressors in zebrafish requires hypothalamic-pituitary-interrenal (HPI) axis activation. In teleost fish, interrenal cells are functionally homologous to the adrenocortical layer. We derived eight frameshift mutants in genes involved in HPI axis function: two mutants in exon 2 of mc2r (adrenocorticotropic hormone receptor), five in exon 2 or 5 of nr3c1 (glucocorticoid receptor [GR]) and two in exon 2 of nr3c2 (mineralocorticoid receptor [MR]). Exposing larval zebrafish to mild environmental stressors, acute changes in salinity or light illumination, results in a rapid locomotor response. We show that this locomotor response requires a functioning HPI axis via the action of mc2r and the canonical GR encoded by nr3c1 gene, but not MR (nr3c2). Our rapid behavioral assay paradigm based on HPI axis biology can be used to screen for genetic and environmental modifiers of the hypothalamic-pituitary-adrenal axis and to investigate the effects of corticosteroids and their cognate receptor interactions on behavior.
Collapse
Affiliation(s)
- Han B. Lee
- Neuroscience Graduate ProgramMayo Clinic Graduate School of Biomedical SciencesRochesterMinnesota
| | - Tanya L. Schwab
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Ashley N. Sigafoos
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Jennifer L. Gauerke
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Randall G. Krug
- Neuroscience Graduate ProgramMayo Clinic Graduate School of Biomedical SciencesRochesterMinnesota
| | - MaKayla R. Serres
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Dakota C. Jacobs
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Ryan P. Cotter
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Biswadeep Das
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Morgan O. Petersen
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Camden L. Daby
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Rhianna M. Urban
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Bethany C. Berry
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| | - Karl J. Clark
- Neuroscience Graduate ProgramMayo Clinic Graduate School of Biomedical SciencesRochesterMinnesota
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesota
| |
Collapse
|