1
|
Leskinen HL, Udvadia AJ. Development and Validation of a Proximity Labeling Fusion Protein Construct to Identify the Protein-Protein Interactions of Transcription Factors. Methods Mol Biol 2025; 2848:269-297. [PMID: 39240529 DOI: 10.1007/978-1-0716-4087-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Dynamic interactions between transcription factors govern changes in gene expression that mediate changes in cell state accompanying injury response and regeneration. Transcription factors frequently function as obligate dimers whose activity is often modulated by post-translational modifications. These critical and often transient interactions are not easily detected by traditional methods to investigate protein-protein interactions. This chapter discusses the design and validation of a fusion protein involving a transcription factor tethered to a proximity labeling ligase, APEX2. In this technique, proteins are biotinylated within a small radius of the transcription factor of interest, regardless of time of interaction. Here we discuss the validations required to ensure proper functioning of the transcription factor proximity labeling tool and the sample preparation of biotinylated proteins for mass spectrometry analysis of putative protein interactors.
Collapse
Affiliation(s)
- Heather L Leskinen
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ava J Udvadia
- Department of Biology, Appalachian State University, Boone, NC, USA.
| |
Collapse
|
2
|
Hu M, Veldman MB. Intraocular Axon Regeneration in a Model of Penetrating Eye Injury. J Ocul Pharmacol Ther 2023; 39:563-571. [PMID: 37486664 PMCID: PMC10616938 DOI: 10.1089/jop.2023.0048] [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: 04/19/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023] Open
Abstract
Purpose: Penetrating eye injuries commonly cause permanent loss of vision in patients. Unlike mammals, zebrafish can regenerate both damaged tissue and severed axons in the central nervous system. Here, we present a tractable adult zebrafish model to study intraocular axon regeneration after penetrating eye injury. Methods: To create consistent penetrating intraocular injuries, pins of standardized diameters were inserted into the eye through the cornea and penetrating the retina but not the underlying sclera. Transgenic gap43:GFP reporter fish were used to preferentially label retinal ganglion cells (RGCs) that respond to injury with regenerating axons. Retinas were fixed and flat mounted at various times postinjury to examine injury size, number of green fluorescent protein (GFP)-positive cells and axons, axonal varicosities, and rate of regeneration to the optic nerve head. Intraocular injection of colchicine was used to inhibit axon outgrow as a proof of principle that this method can be used to screen effects of pharmacological agents on intraocular axon regeneration. Results: Penetrating injury to the zebrafish retina results in robust axon regeneration by RGCs around and beyond the site of injury. The gap43:GFP transgene allows visualization of individual or small bundles of axons with varicosities and growth cones easily observable. Regeneration proceeded with most, if not all, axons reaching the optic nerve head by 3-day postinjury. A single intraocular injection of colchicine a day after injury was sufficient to delay axon regeneration at 2-days postinjury. Surprisingly, we identified a stereotypically located population of circumferential projecting neurons within the retina that upregulate gap43:GFP expression after injury. Conclusions: Penetrating injury to the adult gap43:GFP transgenic zebrafish eye is a model of successful intraocular axon regeneration. The pharmacological and genetic tools available for this organism should make it a powerful tool for dissecting the cellular, molecular, and genetic mechanisms of axon regeneration in the intraocular environment.
Collapse
Affiliation(s)
- Mengming Hu
- Department of Cell Biology, Neurobiology, and Anatomy and Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Matthew B. Veldman
- Department of Cell Biology, Neurobiology, and Anatomy and Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
3
|
Kawasaki T, Fujimori KE, Imada J, Yuba S. Analysis of medaka GAP43 gene promoter activity in transgenic lines. Gene 2023:147590. [PMID: 37364694 DOI: 10.1016/j.gene.2023.147590] [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: 02/20/2023] [Revised: 06/03/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
We produced transgenic medaka fish lines that mimicked the expression of the GAP43 gene. Fish lines with the proximal 2-kilobase (kb) 5'-untranslated region (UTR) as the expression promoter specifically expressed enhanced green fluorescent protein (EGFP) in neural tissues, such as the brain, spinal cord, and peripheral nerves, and its expression decreased with growth, but persisted until adulthood. A functional analysis of the promoter using partially deleted UTRs revealed that functions related to neural tissue-specific promoter activity were widely distributed in the region upstream of the proximal 400-b. Furthermore, the distal half of the 2-kb UTR contributed to expression throughout the brain, while the region 400-b upstream of the proximal 600-b was strongly associated with expression in specific areas, such as the telencephalon. In addition, a region from 957 to 557 b upstream of the translation initiation site was important for the long-term maintenance of promoter activity into adulthood. Among the transcription factors with recognition sequences in this region, Sp1 and CREB1 have been suggested to play important roles in the GAP43 promoter expression characteristics, such as strong expression in the telencephalon and long-term maintenance of expression.
Collapse
Affiliation(s)
- Takashi Kawasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| | - Kazuhiro E Fujimori
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba, Ibaraki 305-0046, Japan.
| | - Junko Imada
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| | - Shunsuke Yuba
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Midorigaoka, Ikeda, Osaka, 563-8577, Japan.
| |
Collapse
|
4
|
Van Dyck A, Masin L, Bergmans S, Schevenels G, Beckers A, Vanhollebeke B, Moons L. A new microfluidic model to study dendritic remodeling and mitochondrial dynamics during axonal regeneration of adult zebrafish retinal neurons. Front Mol Neurosci 2023; 16:1196504. [PMID: 37396787 PMCID: PMC10307971 DOI: 10.3389/fnmol.2023.1196504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/10/2023] [Indexed: 07/04/2023] Open
Abstract
Unlike mammals, adult zebrafish are able to fully regenerate axons and functionally recover from neuronal damage in the mature central nervous system (CNS). Decades of research have tried to identify the mechanisms behind their spontaneous regenerative capacity, but the exact underlying pathways and molecular drivers remain to be fully elucidated. By studying optic nerve injury-induced axonal regrowth of adult zebrafish retinal ganglion cells (RGCs), we previously reported transient dendritic shrinkage and changes in the distribution and morphology of mitochondria in the different neuronal compartments throughout the regenerative process. These data suggest that dendrite remodeling and temporary changes in mitochondrial dynamics contribute to effective axonal and dendritic repair upon optic nerve injury. To further elucidate these interactions, we here present a novel adult zebrafish microfluidic model in which we can demonstrate compartment-specific alterations in resource allocation in real-time at single neuron level. First, we developed a pioneering method that enables to isolate and culture adult zebrafish retinal neurons in a microfluidic setup. Notably, with this protocol, we report on a long-term adult primary neuronal culture with a high number of surviving and spontaneously outgrowing mature neurons, which was thus far only very limitedly described in literature. By performing time-lapse live cell imaging and kymographic analyses in this setup, we can explore changes in dendritic remodeling and mitochondrial motility during spontaneous axonal regeneration. This innovative model system will enable to discover how redirecting intraneuronal energy resources supports successful regeneration in the adult zebrafish CNS, and might facilitate the discovery of new therapeutic targets to promote neuronal repair in humans.
Collapse
Affiliation(s)
- Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Giel Schevenels
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Meehan SD, Hu M, Veldman MB, Bhattacharya SK. Metabolomics dataset of zebrafish optic nerve regeneration after injury. Data Brief 2023; 48:109102. [PMID: 37383800 PMCID: PMC10293924 DOI: 10.1016/j.dib.2023.109102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 06/30/2023] Open
Abstract
Zebrafish (Danio rerio) have the capacity for successful adult optic nerve regeneration. In contrast, mammals lack this intrinsic ability and undergo irreversible neurodegeneration seen in glaucoma and other optic neuropathies. Optic nerve regeneration is often studied using optic nerve crush, a mechanical neurodegenerative model. Untargeted metabolomic studies within successful regenerative models are deficient. Evaluation of tissue metabolomic changes in active zebrafish optic nerve regeneration can elucidate prioritized metabolite pathways that can be targeted in mammalian systems for therapeutic development. Female and male (6 month to 1 year old wild type) right zebrafish optic nerves were crushed and collected three days after. Contralateral, uninjured optic nerves were collected as controls. The tissue was dissected from euthanized fish and frozen on dry ice. Samples were pooled for each category (female crush, female control, male crush, male control) and pooled at n = 31 to obtain sufficient metabolite concentrations for analysis. Optic nerve regeneration at 3 days post crush was demonstrated by microscope visualization of GFP fluorescence in Tg(gap43:GFP) transgenic fish. Metabolites were extracted using a Precellys Homogenizer and a serial extraction method: (1) 1:1 Methanol/Water and (2) 8:1:1 Acetonitrile/Methanol/Acetone. Metabolites were analyzed by untargeted liquid chromatography-mass spectrometry (LC MS-MS) profiling using a Q-Exactive Orbitrap instrument coupled with Vanquish Horizon Binary UHPLC LC-MS system. Metabolites were identified and quantified using Compound Discoverer 3.3 and isotopic internal metabolites standards.
Collapse
Affiliation(s)
- Sean D. Meehan
- Bascom Palmer Eye Institute University of Miami, 1638 NW 10th Avenue, Suite 707A, Miami, FL, 33136, United States
- Miami Integrative Metabolomics Research Center, Miami, FL, 33136, United States
- University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| | - Mengming Hu
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Matthew B. Veldman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, 53226, United States
| | - Sanjoy K. Bhattacharya
- Bascom Palmer Eye Institute University of Miami, 1638 NW 10th Avenue, Suite 707A, Miami, FL, 33136, United States
- Miami Integrative Metabolomics Research Center, Miami, FL, 33136, United States
- University of Miami Miller School of Medicine, Miami, FL, 33136, United States
| |
Collapse
|
6
|
Iyer S, Dhiman N, Zade SP, Mukherjee S, Singla N, Kumar M. Exposure to Tetrabutylammonium Bromide Impairs Cranial Neural Crest Specification, Neurogenic Program, and Brain Morphogenesis. ACS Chem Neurosci 2023; 14:1785-1798. [PMID: 37125651 DOI: 10.1021/acschemneuro.2c00728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Tetrabutylammonium bromide (TBAB) is a widely used industrial reagent and is commonly found in our aquatic ecosystem as an industrial byproduct. In humans, the ingestion of TBAB causes severe neurological impairments and disorders such as vertigo, hallucinations, and delirium. Yet, the extent of environmental risk and TBAB toxicity to human health is poorly understood. In this study, we aim to determine the developmental toxicity of TBAB using zebrafish embryos as a model and provide novel insights into the mechanism of action of such chemicals on neurodevelopment and the overall embryonic program. Our results show that exposure to TBAB results in impaired development of the brain, inner ear, and pharyngeal skeletal elements in the zebrafish embryo. TBAB treatment resulted in aberrations in the specification of the neural crest precursors, hindbrain segmentation, and otic neurogenesis. TBAB treatment also induced a surge in apoptosis in the head, tail, and trunk regions of the developing embryo. Long-term TBAB exposure resulted in cardiac edema and craniofacial defects. Further, in silico molecular docking analysis indicated that TBAB binds to AMPA receptors and modulates neural developmental genes such as olfactomedin and acetylcholinesterase in the embryonic brain. To summarize, our study highlights the novel effects of TBAB on embryonic brain formation and segmentation, ear morphogenesis, and craniofacial skeletal development.
Collapse
Affiliation(s)
- Sharada Iyer
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Uppal Road, Habsiguda, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neha Dhiman
- Department of Biochemistry, Panjab University, Chandigarh160014, India
| | - Suraj P Zade
- Global Product Compliance─India, 301, Samved Sankul, Near MLA Hostel, Civil Lines, Nagpur 440001, India
| | - Sulagna Mukherjee
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Uppal Road, Habsiguda, Hyderabad 500007, India
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh160014, India
| | - Megha Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Uppal Road, Habsiguda, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
7
|
Dhara SP, Udvadia AJ. Profiling Dynamic Changes in DNA Accessibility During Axon Regeneration After Optic Nerve Crush in Adult Zebrafish. Methods Mol Biol 2023; 2636:323-341. [PMID: 36881309 DOI: 10.1007/978-1-0716-3012-9_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
A time-course series utilizing assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) can be used to detect changes in accessibility of DNA regulatory elements such as promoters and enhancers over the course of regeneration. This chapter describes methods for preparing ATAC-seq libraries from isolated zebrafish retinal ganglion cells (RGCs) following optic nerve crush at selected post-injury time points. These methods have been used for identifying dynamic changes in DNA accessibility that govern successful optic nerve regeneration in zebrafish. This method may be adapted to identify changes in DNA accessibility that accompany other types of insults to RGCs or to identify changes that occur over the course of development.
Collapse
Affiliation(s)
- Sumona P Dhara
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ava J Udvadia
- Department of Biology, Appalachian State University, Boone, NC, USA.
| |
Collapse
|
8
|
Analysis of Axonal Regrowth and Dendritic Remodeling After Optic Nerve Crush in Adult Zebrafish. Methods Mol Biol 2023; 2636:163-190. [PMID: 36881300 DOI: 10.1007/978-1-0716-3012-9_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Neurodegenerative diseases and central nervous system (CNS) injuries are frequently characterized by axonal damage, as well as dendritic pathology. In contrast to mammals, adult zebrafish show a robust regeneration capacity after CNS injury and form the ideal model organism to further unravel the underlying mechanisms for both axonal and dendritic regrowth upon CNS damage. Here, we first describe an optic nerve crush injury model in adult zebrafish, an injury paradigm that inflicts de- and regeneration of the axons of retinal ganglion cells (RGCs), but also triggers RGC dendrite disintegration and subsequent recovery in a stereotyped and timed process. Next, we outline protocols for quantifying axonal regeneration and synaptic recovery in the brain, using retro- and anterograde tracing experiments and an immunofluorescent staining for presynaptic compartments, respectively. Finally, methods to analyze RGC dendrite retraction and subsequent regrowth in the retina are delineated, using morphological measurements and immunofluorescent staining for dendritic and synaptic markers.
Collapse
|
9
|
Ferre-Fernández JJ, Muheisen S, Thompson S, Semina EV. CRISPR-Cas9-mediated functional dissection of the foxc1 genomic region in zebrafish identifies critical conserved cis-regulatory elements. Hum Genomics 2022; 16:49. [PMID: 36284357 PMCID: PMC9597995 DOI: 10.1186/s40246-022-00423-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
FOXC1 encodes a forkhead-domain transcription factor associated with several ocular disorders. Correct FOXC1 dosage is critical to normal development, yet the mechanisms controlling its expression remain unknown. Together with FOXQ1 and FOXF2, FOXC1 is part of a cluster of FOX genes conserved in vertebrates. CRISPR-Cas9-mediated dissection of genomic sequences surrounding two zebrafish orthologs of FOXC1 was performed. This included five zebrafish-human conserved regions, three downstream of foxc1a and two remotely upstream of foxf2a/foxc1a or foxf2b/foxc1b clusters, as well as two intergenic regions between foxc1a/b and foxf2a/b lacking sequence conservation but positionally corresponding to the area encompassing a previously reported glaucoma-associated SNP in humans. Removal of downstream sequences altered foxc1a expression; moreover, zebrafish carrying deletions of two or three downstream elements demonstrated abnormal phenotypes including enlargement of the anterior chamber of the eye reminiscent of human congenital glaucoma. Deletions of distant upstream conserved elements influenced the expression of foxf2a/b or foxq1a/b but not foxc1a/b within each cluster. Removal of either intergenic sequence reduced foxc1a or foxc1b expression during late development, suggesting a role in transcriptional regulation despite the lack of conservation at the nucleotide level. Further studies of the identified regions in human patients may explain additional individuals with developmental ocular disorders.
Collapse
Affiliation(s)
- Jesús-José Ferre-Fernández
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sanaa Muheisen
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI, 53226, USA
| | - Samuel Thompson
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI, 53226, USA
| | - Elena V Semina
- Department of Pediatrics and Children's Research Institute, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
10
|
Comparative in situ hybridization protocols in zebrafish. Biotechniques 2022; 73:123-130. [PMID: 36065907 PMCID: PMC9490454 DOI: 10.2144/btn-2022-0038] [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] [Indexed: 11/23/2022] Open
Abstract
In situ hybridization is a commonly used technique in molecular biology to assess the temporal and spatial expression of a given gene. As a long and labor-intensive protocol, double in situ hybridization, which detects two genes in series, is challenging and can require a lot of troubleshooting. Optional additives, polyvinyl alcohol and dextran sulfate, were tested in a standard in situ hybridization protocol and several colorimetric stain pairings using double in situ hybridization in zebrafish embryos. Optional additives can improve staining time and reduce nonspecific background. Nitro-blue tetrazolium chloride/5-bromo-4-chloro-3-indolyl phosphate (BCIP) + Fast Red/BCIP was the most effective stain pairing. As a proof-of-concept, this work shows that Cabin1 and atoh1b are expressed in distinct regions of the developing zebrafish brain. A comparison of colorimetric stains and protocols in double in situ hybridization in whole-mount zebrafish embryos.
Collapse
|
11
|
Beckers A, Vanhunsel S, Van Dyck A, Bergmans S, Masin L, Moons L. Injury-induced Autophagy Delays Axonal Regeneration after Optic Nerve Damage in Adult Zebrafish. Neuroscience 2021; 470:52-69. [PMID: 34280491 DOI: 10.1016/j.neuroscience.2021.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/18/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Optic neuropathies comprise a group of disorders in which the axons of retinal ganglion cells (RGCs), the retinal projection neurons conveying visual information to the brain, are damaged. This results in visual impairment or even blindness, which is irreversible as adult mammals lack the capacity to repair or replace injured or lost neurons. Despite intensive research, no efficient treatment to induce axonal regeneration in the central nervous system (CNS) is available yet. Autophagy, the cellular recycling response, was shown repeatedly to be elevated in animal models of optic nerve injury, and both beneficial and detrimental effects have been reported. In this study, we subjected spontaneously regenerating adult zebrafish to optic nerve damage (ONC) and revealed that autophagy is enhanced after optic nerve damage in zebrafish, both in RGC axons and somas, as well as in macroglial cells of the retina, the optic nerve and the visual target areas in the brain. Interestingly, the pattern of the autophagic response in the axons followed the spatiotemporal window of axonal regrowth, which suggests that autophagy is ongoing at the growth cones. Pharmacological inhibition of the recycling pathway resulted in accelerated RGC target reinnervation, possibly linked to increased mechanistic target of rapamycin (mTOR) activity, known to stimulate axonal regrowth. Taken together, these intriguing findings underline that further research is warranted to decipher if modulation of autophagy could be an effective therapeutic method to induce CNS regeneration.
Collapse
Affiliation(s)
- An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Sophie Vanhunsel
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium; Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
12
|
Schnabl J, Litz MPH, Schneider C, PenkoffLidbeck N, Bashiruddin S, Schwartz MS, Alligood K, Devoto SH, Barresi MJF. Characterizing the diverse cells that associate with the developing commissures of the zebrafish forebrain. Dev Neurobiol 2021; 81:671-695. [PMID: 33314626 DOI: 10.1002/dneu.22801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/20/2020] [Accepted: 12/08/2020] [Indexed: 01/02/2023]
Abstract
During embryonic development of bilaterally symmetrical organisms, neurons send axons across the midline at specific points to connect the two halves of the nervous system with a commissure. Little is known about the cells at the midline that facilitate this tightly regulated process. We exploit the conserved process of vertebrate embryonic development in the zebrafish model system to elucidate the identity of cells at the midline that may facilitate postoptic (POC) and anterior commissure (AC) development. We have discovered that three different gfap+ astroglial cell morphologies persist in contact with pathfinding axons throughout commissure formation. Similarly, olig2+ progenitor cells occupy delineated portions of the postoptic and anterior commissures where they act as multipotent, neural progenitors. Moreover, we conclude that both gfap+ and olig2+ progenitor cells give rise to neuronal populations in both the telencephalon and diencephalon; however, these varied cell populations showed significant developmental timing differences between the telencephalon and diencephalon. Lastly, we also showed that fli1a+ mesenchymal cells migrate along the presumptive commissure regions before and during midline axon crossing. Furthermore, following commissure maturation, specific blood vessels formed at the midline of the POC and immediately ventral and parallel to the AC. This comprehensive account of the cellular populations that correlate with the timing and position of commissural axon pathfinding has supported the conceptual modeling and identification of the early forebrain architecture that may be necessary for proper commissure development.
Collapse
Affiliation(s)
- Jake Schnabl
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Mackenzie P H Litz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caitlin Schneider
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,McGill University, Montreal, QC, Canada
| | | | - Sarah Bashiruddin
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Family Medicine Assoc, Westfield, MA, USA
| | - Morgan S Schwartz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kristin Alligood
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Farmers Conservation Alliance, Hood River, OR, USA
| | | | - Michael J F Barresi
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA.,Department of Biological Sciences, Smith College, Northampton, MA, USA
| |
Collapse
|
13
|
Clark EM, Link BA. Complementary and divergent functions of zebrafish Tango1 and Ctage5 in tissue development and homeostasis. Mol Biol Cell 2021; 32:391-401. [PMID: 33439675 PMCID: PMC8098853 DOI: 10.1091/mbc.e20-11-0745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 01/05/2023] Open
Abstract
Coat protein complex II (COPII) factors mediate cargo export from the endoplasmic reticulum (ER), but bulky collagens and lipoproteins are too large for traditional COPII vesicles. Mammalian CTAGE5 and TANGO1 have been well characterized individually as specialized cargo receptors at the ER that function with COPII coats to facilitate trafficking of bulky cargoes. Here, we present a genetic interaction study in zebrafish of deletions in ctage5, tango1, or both to investigate their distinct and complementary potential functions. We found that Ctage5 and Tango1 have different roles related to organogenesis, collagen versus lipoprotein trafficking, stress-pathway activation, and survival. While disruption of both ctage5 and tango1 compounded phenotype severity, mutation of either factor alone revealed novel tissue-specific defects in the building of heart, muscle, lens, and intestine, in addition to previously described roles in the development of neural and cartilage tissues. Together, our results demonstrate that Ctage5 and Tango1 have overlapping functions, but also suggest divergent roles in tissue development and homeostasis.
Collapse
Affiliation(s)
- Eric M. Clark
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Brian A. Link
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
14
|
Qian L, Qi S, Wang Z, Magnuson JT, Volz DC, Schlenk D, Jiang J, Wang C. Environmentally relevant concentrations of boscalid exposure affects the neurobehavioral response of zebrafish by disrupting visual and nervous systems. JOURNAL OF HAZARDOUS MATERIALS 2021; 404:124083. [PMID: 33011634 DOI: 10.1016/j.jhazmat.2020.124083] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/02/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
Boscalid is a persistent fungicide that is frequently detected in surface waters and may be neurotoxic to aquatic organisms. Herein, we evaluated the effects of environmentally relevant boscalid concentrations to zebrafish to explore its potentially neurotoxic mechanisms of effect. Behavioral responses (swimming, phototaxis, and predation), histopathology, transcriptomics, biochemical parameter analysis and gene expression of larval and adult zebrafish following boscalid treatment were assessed. We found that boscalid significantly inhibited the locomotor ability and phototactic response of larvae after an 8-d exposure, and altered the locomotor activity, predation trajectories and ability in adults after a 21-d exposure. It was noted that predation rates of zebrafish were significantly decreased by 30% and 100% after exposure to 0.1 and 1.0 mg/L boscalid, respectively. Adverse alterations in the cell differentiation of eyes and brain injury were also observed in both larvae and adults following boscalid exposure. The expression of genes related to neurodevelopment, neurotransmission, eye development, and visual function, in conjunction with RNA-Seq results, indicated that boscalid may impair visual phototransduction and nervous system processes in larval zebrafish. Conclusively, boscalid exposure may affect the neurobehavioral response of zebrafish by impairing proper visual and nervous system function.
Collapse
Affiliation(s)
- Le Qian
- College of Sciences, China Agricultural University, Beijing, China
| | - Suzhen Qi
- Risk Assessment Laboratory for Bee Products Quality and Safety of Ministry of Agriculture, Institute of Agricultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhao Wang
- Institute of Plant Protection, Jilin Academy of Agricultural Sciences, China
| | - Jason T Magnuson
- Department of Environmental Sciences, University of California, Riverside, Riverside, CA, United States
| | - David C Volz
- Department of Environmental Sciences, University of California, Riverside, Riverside, CA, United States
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, Riverside, CA, United States
| | - Jiazhen Jiang
- College of Sciences, China Agricultural University, Beijing, China.
| | - Chengju Wang
- College of Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
15
|
Naderi M, Kwong RWM. A comprehensive review of the neurobehavioral effects of bisphenol S and the mechanisms of action: New insights from in vitro and in vivo models. ENVIRONMENT INTERNATIONAL 2020; 145:106078. [PMID: 32911243 DOI: 10.1016/j.envint.2020.106078] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
The normal brain development and function are delicately driven by an ever-changing milieu of steroid hormones arising from fetal, placental, and maternal origins. This reliance on the neuroendocrine system sets the stage for the exquisite sensitivity of the central nervous system to the adverse effects of endocrine-disrupting chemicals (EDCs). Bisphenol A (BPA) is one of the most common EDCs which has been a particular focus of environmental concern for decades due to its widespread nature and formidable threat to human and animal health. The heightened regulatory actions and the scientific and public concern over the adverse health effects of BPA have led to its replacement with a suite of structurally similar but less known alternative chemicals. Bisphenol S (BPS) is the main substitute for BPA that is increasingly being used in a wide array of consumer and industrial products. Although it was considered to be a safe BPA alternative, mounting evidence points to the deleterious effects of BPS on a wide range of neuroendocrine functions in animals. In addition to its reproductive toxicity, recent experimental efforts indicate that BPS has a considerable potential to induce neurotoxicity and behavioral dysfunction. This review analyzes the current state of knowledge regarding the neurobehavioral effects of BPS and discusses its potential mode of actions on several aspects of the neuroendocrine system. We summarize the role of certain hormones and their signaling pathways in the regulation of brain and behavior and discuss how BPS induces neurotoxicity through interactions with these pathways. Finally, we review potential links between BPS exposure and aberrant neurobehavioral functions in animals and identify key knowledge gaps and hypotheses for future research.
Collapse
Affiliation(s)
- Mohammad Naderi
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| | - Raymond W M Kwong
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
16
|
Dhara SP, Rau A, Flister MJ, Recka NM, Laiosa MD, Auer PL, Udvadia AJ. Cellular reprogramming for successful CNS axon regeneration is driven by a temporally changing cast of transcription factors. Sci Rep 2019; 9:14198. [PMID: 31578350 PMCID: PMC6775158 DOI: 10.1038/s41598-019-50485-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/10/2019] [Indexed: 11/10/2022] Open
Abstract
In contrast to mammals, adult fish display a remarkable ability to fully regenerate central nervous system (CNS) axons, enabling functional recovery from CNS injury. Both fish and mammals normally undergo a developmental downregulation of axon growth activity as neurons mature. Fish are able to undergo damage-induced “reprogramming” through re-expression of genes necessary for axon growth and guidance, however, the gene regulatory mechanisms remain unknown. Here we present the first comprehensive analysis of gene regulatory reprogramming in zebrafish retinal ganglion cells at specific time points along the axon regeneration continuum from early growth to target re-innervation. Our analyses reveal a regeneration program characterized by sequential activation of stage-specific pathways, regulated by a temporally changing cast of transcription factors that bind to stably accessible DNA regulatory regions. Strikingly, we also find a discrete set of regulatory regions that change in accessibility, consistent with higher-order changes in chromatin organization that mark (1) the beginning of regenerative axon growth in the optic nerve, and (2) the re-establishment of synaptic connections in the brain. Together, these data provide valuable insight into the regulatory logic driving successful vertebrate CNS axon regeneration, revealing key gene regulatory candidates for therapeutic development.
Collapse
Affiliation(s)
- Sumona P Dhara
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Andrea Rau
- GABI, INRA, AgroParisTech, Universite Paris-Saclay, 78350, Jouy-en-Josas, France.,Joseph J Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Michael J Flister
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Nicole M Recka
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Michael D Laiosa
- Joseph J Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Paul L Auer
- Joseph J Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Ava J Udvadia
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA.
| |
Collapse
|
17
|
Daniel JG, Panizzi JR. Spatiotemporal expression profile of embryonic and adult ankyrin repeat and EF-hand domain containing protein 1-encoding genes ankef1a and ankef1b in zebrafish. Gene Expr Patterns 2019; 34:119069. [PMID: 31520739 DOI: 10.1016/j.gep.2019.119069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Recent human next-generation sequencing (NGS) studies indicate a correlation between ANKEF1 (ankyrin repeat and EF-hand domain containing protein 1) expression and cilia formation or function. Additionally, a single study conducted in the African clawed frog (Xenopus laevis) showed ankef1 is down-regulated after pharmacological fibroblast growth factor (FGF) inhibition and plays a role in protocadherin-mediated cell protrusion and adhesion. That study also revealed a critical role for ankef1 in the embryonic development of the frog, with morphants exhibiting phenotypes including spina bifida and a shortened body axis. Interestingly, while little is known about ANKEF1 function in other vertebrate systems, recent proteomic data has shown ANKEF1 enriched in ciliated cells. Likewise, publicly available EST profile databases imply ANKEF1 expression in multiple human tissues, including high levels in the testes. Together, these previous studies suggest an important role for ANKEF1 in ciliated tissues and during embryonic development. Here, we report cloning of zebrafish (Danio rerio) ankef1a, as well as its paralog, ankef1b, and expression analyses by whole-mount in situ hybridization (WISH) and quantitative polymerase chain reaction (qPCR) during embryonic development and in adult tissues. WISH shows both forms are ubiquitously expressed early in development, with more discrete expression of both transcripts in embryonic tissues known to precede or possess motile cilia, including dorsal forerunner cells (DFC) and the otic vesicles, respectively. Additionally, both transcripts are enriched in the developing pharynx and swim bladder. Our qPCR results indicate enhanced expression in the testes, along with increased expression in brain. Certainly, our experiments in the zebrafish model system with ankef1a and ankef1b provide a solid foundation for future studies to uncover the molecular pathways through which Ankef1 acts in both healthy and disease states.
Collapse
Affiliation(s)
- Jeffrey G Daniel
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, 36849, USA
| | - Jennifer R Panizzi
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, 36849, USA.
| |
Collapse
|
18
|
Das S, Sreevidya VS, Udvadia AJ, Gyaneshwar P. Dopamine-induced sulfatase and its regulator are required for Salmonella enterica serovar Typhimurium pathogenesis. MICROBIOLOGY-SGM 2019; 165:302-310. [PMID: 30648943 DOI: 10.1099/mic.0.000769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Catecholamine hormones enhance the virulence of pathogenic bacteria. Studies in the 1980s made intriguing observations that catecholamines were required for induction of sulfatase activity in many enteric pathogens, including Salmonella enterica serovar Typhimurium. In this report, we show that STM3122 and STM3124, part of horizontally acquired Salmonella pathogenesis island 13, encode a catecholamine-induced sulfatase and its regulator, respectively. Induction of sulfatase activity was independent of the well-studied QseBC and QseEF two-component regulatory systems. S. Typhimurium 14028S mutants lacking STM3122 or STM3124 showed reduced virulence in zebrafish. Because catecholamines are inactivated by sulfation in the mammalian gut, S. Typhimurium could utilize CA-induced sulfatase to access free catecholamines for growth and virulence.
Collapse
Affiliation(s)
- Seema Das
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | | | - Ava J Udvadia
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Prasad Gyaneshwar
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
19
|
Beckers A, Van Dyck A, Bollaerts I, Van houcke J, Lefevere E, Andries L, Agostinone J, Van Hove I, Di Polo A, Lemmens K, Moons L. An Antagonistic Axon-Dendrite Interplay Enables Efficient Neuronal Repair in the Adult Zebrafish Central Nervous System. Mol Neurobiol 2018; 56:3175-3192. [DOI: 10.1007/s12035-018-1292-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/31/2018] [Indexed: 11/29/2022]
|
20
|
Fish Scales Dictate the Pattern of Adult Skin Innervation and Vascularization. Dev Cell 2018; 46:344-359.e4. [PMID: 30032992 DOI: 10.1016/j.devcel.2018.06.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 05/27/2018] [Accepted: 06/22/2018] [Indexed: 11/24/2022]
Abstract
As animals mature from embryonic to adult stages, the skin grows and acquires specialized appendages, like hairs, feathers, and scales. How cutaneous blood vessels and sensory axons adapt to these dramatic changes is poorly understood. By characterizing skin maturation in zebrafish, we discovered that sensory axons are delivered to the adult epidermis in organized nerves patterned by features in bony scales. These nerves associate with blood vessels and osteoblasts above scales. Osteoblasts create paths in scales that independently guide nerves and blood vessels during both development and regeneration. By preventing scale regeneration and examining mutants lacking scales, we found that scales recruit, organize, and polarize axons and blood vessels to evenly distribute them in the skin. These studies uncover mechanisms for achieving comprehensive innervation and vascularization of the adult skin and suggest that scales coordinate a metamorphosis-like transformation of the skin with sensory axon and vascular remodeling.
Collapse
|
21
|
Green AJ, Planchart A. The neurological toxicity of heavy metals: A fish perspective. Comp Biochem Physiol C Toxicol Pharmacol 2018; 208:12-19. [PMID: 29199130 PMCID: PMC5936656 DOI: 10.1016/j.cbpc.2017.11.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022]
Abstract
The causes of neurodegenerative diseases are complex with likely contributions from genetic susceptibility and environmental exposures over an organism's lifetime. In this review, we examine the role that aquatic models, especially zebrafish, have played in the elucidation of mechanisms of heavy metal toxicity and nervous system function over the last decade. Focus is applied to cadmium, lead, and mercury as significant contributors to central nervous system morbidity, and the application of numerous transgenic zebrafish expressing fluorescent reporters in specific neuronal populations or brain regions enabling high-resolution neurodevelopmental and neurotoxicology research.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Behavior, Animal/drug effects
- Disease Models, Animal
- Gene Expression Regulation, Developmental/drug effects
- Heavy Metal Poisoning, Nervous System/etiology
- Heavy Metal Poisoning, Nervous System/genetics
- Heavy Metal Poisoning, Nervous System/metabolism
- Heavy Metal Poisoning, Nervous System/pathology
- Humans
- Metals, Heavy/toxicity
- Nerve Degeneration
- Nervous System/drug effects
- Nervous System/metabolism
- Nervous System/pathology
- Nervous System/physiopathology
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
- Risk Assessment
- Water Pollutants, Chemical/toxicity
- Zebrafish/genetics
- Zebrafish/metabolism
Collapse
Affiliation(s)
- Adrian J Green
- Graduate Program in Toxicology, North Carolina State University, Raleigh, NC 27695, United States; Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States
| | - Antonio Planchart
- Graduate Program in Toxicology, North Carolina State University, Raleigh, NC 27695, United States; Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, United States; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, United States; W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, United States.
| |
Collapse
|
22
|
Bollaerts I, Veys L, Geeraerts E, Andries L, De Groef L, Buyens T, Salinas-Navarro M, Moons L, Van Hove I. Complementary research models and methods to study axonal regeneration in the vertebrate retinofugal system. Brain Struct Funct 2017; 223:545-567. [DOI: 10.1007/s00429-017-1571-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/15/2017] [Indexed: 01/18/2023]
|
23
|
Learning to swim, again: Axon regeneration in fish. Exp Neurol 2017; 287:318-330. [DOI: 10.1016/j.expneurol.2016.02.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/25/2016] [Accepted: 02/27/2016] [Indexed: 01/10/2023]
|
24
|
Levin E, Diekmann H, Fischer D. Highly efficient transduction of primary adult CNS and PNS neurons. Sci Rep 2016; 6:38928. [PMID: 27958330 PMCID: PMC5153636 DOI: 10.1038/srep38928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/15/2016] [Indexed: 12/19/2022] Open
Abstract
Delivery and expression of recombinant genes, a key methodology for many applications in biological research, remains a challenge especially for mature neurons. Here, we report easy, highly efficient and well tolerated transduction of adult peripheral and central neuronal populations of diverse species in culture using VSV-G pseudo-typed, recombinant baculovirus (BacMam). Transduction rates of up to 80% were reliably achieved at high multiplicity of infection without apparent neuro-cytopathic effects. Neurons could be transduced either shortly after plating or after several days in culture. Co-incubation with two different baculoviruses attained near complete co-localization of fluorescent protein expression, indicating multigene delivery. Finally, evidence for functional protein expression is provided by means of cre-mediated genetic recombination and neurite outgrowth assays. Recombinant protein was already detected within hours after transduction, thereby enabling functional readouts even in relatively short-lived neuronal cultures. Altogether, these results substantiate the usefulness of baculovirus-mediated transduction of mature neurons for future research in neuroscience.
Collapse
Affiliation(s)
- Evgeny Levin
- Division of Experimental Neurology, Medical Faculty, Heinrich-Heine-University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Heike Diekmann
- Division of Experimental Neurology, Medical Faculty, Heinrich-Heine-University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| | - Dietmar Fischer
- Division of Experimental Neurology, Medical Faculty, Heinrich-Heine-University, Merowingerplatz 1a, 40225 Düsseldorf, Germany
| |
Collapse
|
25
|
Liu Z, Wang Y, Zhu Z, Yang E, Feng X, Fu Z, Jin Y. Atrazine and its main metabolites alter the locomotor activity of larval zebrafish (Danio rerio). CHEMOSPHERE 2016; 148:163-170. [PMID: 26803580 DOI: 10.1016/j.chemosphere.2016.01.007] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/03/2016] [Accepted: 01/04/2016] [Indexed: 06/05/2023]
Abstract
Atrazine (ATZ) and its main chlorometabolites, i.e., diaminochlorotriazine (DACT), deisopropylatrazine (DIP), and deethylatrazine (DE), have been widely detected in aquatic systems near agricultural fields. However, their possible effects on aquatic animals are still not fully understood. In this study, it was observed that several developmental endpoints such as the heart beat, hatchability, and morphological abnormalities were influenced by ATZ and its metabolites in different developmental stages. In addition, after 5 days of exposure to 30, 100, 300 μg L(-1) ATZ and its main chlorometabolites, the swimming behaviors of larval zebrafish were significantly disturbed, and the acetylcholinesterase (AChE) activities were consistently inhibited. Our results also demonstrate that ATZ and its main chlorometabolites are neuroendocrine disruptors that impact the expression of neurotoxicity-related genes such as Ache, Gap43, Gfap, Syn2a, Shha, Mbp, Elavl3, Nestin and Ngn1 in early developmental stages of zebrafish. According to our results, it is possible that not only ATZ but also its metabolites (DACT, DIP and DE) have the same or even more toxic effects on different endpoints of the early developmental stages of zebrafish.
Collapse
Affiliation(s)
- Zhenzhen Liu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yueyi Wang
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhihong Zhu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Enlu Yang
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xiayan Feng
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhengwei Fu
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
26
|
Diekmann H, Kalbhen P, Fischer D. Active mechanistic target of rapamycin plays an ancillary rather than essential role in zebrafish CNS axon regeneration. Front Cell Neurosci 2015. [PMID: 26217179 PMCID: PMC4493654 DOI: 10.3389/fncel.2015.00251] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The developmental decrease of the intrinsic regenerative ability of the mammalian central nervous system (CNS) is associated with reduced activity of mechanistic target of rapamycin (mTOR) in mature neurons such as retinal ganglion cells (RGCs). While mTOR activity is further decreased upon axonal injury, maintenance of its pre-injury level, for instance by genetic deletion of the phosphatase and tensin homolog (PTEN), markedly promotes axon regeneration in mammals. The current study now addressed the question whether active mTOR might generally play a central role in axon regeneration by analyzing its requirement in regeneration-competent zebrafish. Remarkably, regulation of mTOR activity after optic nerve injury in zebrafish is fundamentally different compared to mammals. Hardly any activity was detected in naïve RGCs, whereas it was markedly increased upon axotomy in vivo as well as in dissociated cell cultures. After a short burst, mTOR activity was quickly attenuated, which is contrary to the requirements for axon regeneration in mammals. Surprisingly, mTOR activity was not essential for axonal growth per se, but correlated with cytokine- and PTEN inhibitor-induced neurite extension in vitro. Moreover, inhibition of mTOR using rapamycin significantly reduced axon regeneration in vivo and compromised functional recovery after optic nerve injury. Therefore, axotomy-induced mTOR activity is involved in CNS axon regeneration in zebrafish similar to mammals, although it plays an ancillary rather than essential role in this regeneration-competent species.
Collapse
Affiliation(s)
- Heike Diekmann
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University of Düsseldorf Düsseldorf, Germany
| | - Pascal Kalbhen
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University of Düsseldorf Düsseldorf, Germany
| | - Dietmar Fischer
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University of Düsseldorf Düsseldorf, Germany
| |
Collapse
|
27
|
Diekmann H, Kalbhen P, Fischer D. Characterization of optic nerve regeneration using transgenic zebrafish. Front Cell Neurosci 2015; 9:118. [PMID: 25914619 PMCID: PMC4391235 DOI: 10.3389/fncel.2015.00118] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/16/2015] [Indexed: 11/13/2022] Open
Abstract
In contrast to the adult mammalian central nervous system (CNS), fish are able to functionally regenerate severed axons upon injury. Although the zebrafish is a well-established model vertebrate for genetic and developmental studies, its use for anatomical studies of axon regeneration has been hampered by the paucity of appropriate tools to visualize re-growing axons in the adult CNS. On this account, we used transgenic zebrafish that express enhanced green fluorescent protein (GFP) under the control of a GAP-43 promoter. In adult, naïve retinae, GFP was restricted to young retinal ganglion cells (RGCs) and their axons. Within the optic nerve, these fluorescent axons congregated in a distinct strand at the nerve periphery, indicating age-related order. Upon optic nerve crush, GFP expression was markedly induced in RGC somata and intra-retinal axons at 4 to at least 14 days post injury. Moreover, individual axons were visualized in their natural environment of the optic nerve using wholemount tissue clearing and confocal microscopy. With this novel approach, regenerating axons were clearly detectable beyond the injury site as early as 2 days after injury and grew past the optic chiasm by 4 days. Regenerating axons in the entire optic nerve were labeled from 6 to at least 14 days after injury, thereby allowing detailed visualization of the complete regeneration process. Therefore, this new approach could now be used in combination with expression knockdown or pharmacological manipulations to analyze the relevance of specific proteins and signaling cascades for axonal regeneration in vivo. In addition, the RGC-specific GFP expression facilitated accurate evaluation of neurite growth in dissociated retinal cultures. This fast in vitro assay now enables the screening of compound and expression libraries. Overall, the presented methodologies provide exciting possibilities to investigate the molecular mechanisms underlying successful CNS regeneration in zebrafish.
Collapse
Affiliation(s)
- Heike Diekmann
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University of Düsseldorf Düsseldorf, Germany
| | - Pascal Kalbhen
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University of Düsseldorf Düsseldorf, Germany
| | - Dietmar Fischer
- Division of Experimental Neurology, Department of Neurology, Heinrich-Heine-University of Düsseldorf Düsseldorf, Germany
| |
Collapse
|
28
|
Williams RR, Venkatesh I, Pearse DD, Udvadia AJ, Bunge MB. MASH1/Ascl1a leads to GAP43 expression and axon regeneration in the adult CNS. PLoS One 2015; 10:e0118918. [PMID: 25751153 PMCID: PMC4353704 DOI: 10.1371/journal.pone.0118918] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
Unlike CNS neurons in adult mammals, neurons in fish and embryonic mammals can regenerate their axons after injury. These divergent regenerative responses are in part mediated by the growth-associated expression of select transcription factors. The basic helix-loop-helix (bHLH) transcription factor, MASH1/Ascl1a, is transiently expressed during the development of many neuronal subtypes and regulates the expression of genes that mediate cell fate determination and differentiation. In the adult zebrafish (Danio rerio), Ascl1a is also transiently expressed in retinal ganglion cells (RGCs) that regenerate axons after optic nerve crush. Utilizing transgenic zebrafish with a 3.6 kb GAP43 promoter that drives expression of an enhanced green fluorescent protein (EGFP), we observed that knock-down of Ascl1a expression reduces both regenerative gap43 gene expression and axonal growth after injury compared to controls. In mammals, the development of noradrenergic brainstem neurons requires MASH1 expression. In contrast to zebrafish RGCs, however, MASH1 is not expressed in the mammalian brainstem after spinal cord injury (SCI). Therefore, we utilized adeno-associated viral (AAV) vectors to overexpress MASH1 in four month old rat (Rattus norvegicus) brainstem neurons in an attempt to promote axon regeneration after SCI. We discovered that after complete transection of the thoracic spinal cord and implantation of a Schwann cell bridge, animals that express MASH1 exhibit increased noradrenergic axon regeneration and improvement in hindlimb joint movements compared to controls. Together these data demonstrate that MASH1/Ascl1a is a fundamental regulator of axonal growth across vertebrates and can induce modifications to the intrinsic state of neurons to promote functional regeneration in response to CNS injury.
Collapse
Affiliation(s)
- Ryan R. Williams
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Ishwariya Venkatesh
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States of America
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Ava J. Udvadia
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States of America
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
29
|
Bai Q, Parris RS, Burton EA. Different mechanisms regulate expression of zebrafish myelin protein zero (P0) in myelinating oligodendrocytes and its induction following axonal injury. J Biol Chem 2014; 289:24114-28. [PMID: 25028515 DOI: 10.1074/jbc.m113.545426] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Zebrafish CNS axons regenerate robustly following injury; it is thought that CNS oligodendrocytes contribute to this response by expressing growth-promoting molecules. We characterized the mpz gene, which encodes myelin protein zero and is up-regulated in oligodendroglia following axonal injury. The 2.5-kb mpz mRNA is expressed from a single TATA box promoter. Four independent Tg(mpz:egfp) transgenic zebrafish lines, in which GFP was expressed under the mpz promoter and 10 kb of genomic 5'-flanking sequence, showed transgene expression in CNS oligodendrocytes from larval development through adulthood. Following optic nerve crush injury, the mpz:egfp transgene was strongly up-regulated in oligodendrocytes along the regenerating retinotectal projection, mirroring up-regulation of endogenous mpz mRNA. GFP-expressing oligodendroglia were significantly more abundant in the regenerating optic pathway, resulting from both transgene induction in oligodendroglial precursors and the birth of new cells. Up-regulation of the mpz:egfp transgene was not dependent on axonal regeneration, suggesting that the primary signal may be axonal loss, debris, or microglial infiltration. Deletion experiments indicated that an oligodendroglial enhancer located in the region from -6 to -10 kb with respect to the mpz transcriptional start site is dissociable from the cis-regulatory element mediating the mpz transcriptional response to axonal injury, which is located between -1 and -4 kb. These data show that different mechanisms regulate expression of zebrafish mpz in myelinating oligodendrocytes and its induction following axonal injury. The underlying molecular events could potentially be exploited to enhance axonal repair following mammalian CNS injury. The transgenic lines and cis-regulatory constructs reported here will facilitate identification of the relevant signaling pathways.
Collapse
Affiliation(s)
- Qing Bai
- From the Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, and
| | - Ritika S Parris
- From the Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, and
| | - Edward A Burton
- From the Pittsburgh Institute for Neurodegenerative Diseases, Department of Neurology, and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15213 and the Geriatric Research Education and Clinical Center and Department of Neurology, Pittsburgh Veterans Affairs Healthcare System, Pittsburgh, Pennsylvania 15240
| |
Collapse
|
30
|
Sherpa T, Lankford T, McGinn TE, Hunter SS, Frey RA, Sun C, Ryan M, Robison BD, Stenkamp DL. Retinal regeneration is facilitated by the presence of surviving neurons. Dev Neurobiol 2014; 74:851-76. [PMID: 24488694 DOI: 10.1002/dneu.22167] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/27/2014] [Accepted: 01/27/2014] [Indexed: 12/22/2022]
Abstract
Teleost fish regenerate their retinas after damage, in contrast to mammals. In zebrafish subjected to an extensive ouabain-induced lesion that destroys all neurons and spares Müller glia, functional recovery and restoration of normal optic nerve head (ONH) diameter take place at 100 days postinjury. Subsequently, regenerated retinas overproduce cells in the retinal ganglion cell (RGC) layer, and the ONH becomes enlarged. Here, we test the hypothesis that a selective injury, which spares photoreceptors and Müller glia, results in faster functional recovery and fewer long-term histological abnormalities. Following this selective retinal damage, recovery of visual function required 60 days, consistent with this hypothesis. In contrast to extensively damaged retinas, selectively damaged retinas showed fewer histological errors and did not overproduce neurons. Extensively damaged retinas had RGC axons that were delayed in pathfinding to the ONH, and showed misrouted axons within the ONH, suggesting that delayed functional recovery following an extensive lesion is related to defects in RGC axons exiting the eye and/or reaching their central targets. The atoh7, fgf8a, Sonic hedgehog (shha), and netrin-1 genes were differentially expressed, and the distribution of hedgehog protein was disrupted after extensive damage as compared with selective damage. Confirming a role for Shh signaling in supporting rapid regeneration, shha(t4) +/- zebrafish showed delayed functional recovery after selective damage. We suggest that surviving retinal neurons provide structural/molecular information to regenerating neurons, and that this patterning mechanism regulates factors such as Shh. These factors in turn control neuronal number, retinal lamination, and RGC axon pathfinding during retinal regeneration.
Collapse
Affiliation(s)
- Tshering Sherpa
- Department of Biological Sciences, University of Idaho, Moscow, Idaho; Department of Biological Sciences, Graduate Program in Neuroscience, University of Idaho, Moscow, Idaho
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Veth KN, Willer JR, Collery RF, Gray MP, Willer GB, Wagner DS, Mullins MC, Udvadia AJ, Smith RS, John SWM, Gregg RG, Link BA. Mutations in zebrafish lrp2 result in adult-onset ocular pathogenesis that models myopia and other risk factors for glaucoma. PLoS Genet 2011; 7:e1001310. [PMID: 21379331 PMCID: PMC3040661 DOI: 10.1371/journal.pgen.1001310] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 01/13/2011] [Indexed: 11/18/2022] Open
Abstract
The glaucomas comprise a genetically complex group of retinal neuropathies that typically occur late in life and are characterized by progressive pathology of the optic nerve head and degeneration of retinal ganglion cells. In addition to age and family history, other significant risk factors for glaucoma include elevated intraocular pressure (IOP) and myopia. The complexity of glaucoma has made it difficult to model in animals, but also challenging to identify responsible genes. We have used zebrafish to identify a genetically complex, recessive mutant that shows risk factors for glaucoma including adult onset severe myopia, elevated IOP, and progressive retinal ganglion cell pathology. Positional cloning and analysis of a non-complementing allele indicated that non-sense mutations in low density lipoprotein receptor-related protein 2 (lrp2) underlie the mutant phenotype. Lrp2, previously named Megalin, functions as an endocytic receptor for a wide-variety of bioactive molecules including Sonic hedgehog, Bone morphogenic protein 4, retinol-binding protein, vitamin D-binding protein, and apolipoprotein E, among others. Detailed phenotype analyses indicated that as lrp2 mutant fish age, many individuals—but not all—develop high IOP and severe myopia with obviously enlarged eye globes. This results in retinal stretch and prolonged stress to retinal ganglion cells, which ultimately show signs of pathogenesis. Our studies implicate altered Lrp2-mediated homeostasis as important for myopia and other risk factors for glaucoma in humans and establish a new genetic model for further study of phenotypes associated with this disease. Complex genetic inheritance, including variable penetrance and severity, underlies many common eye diseases. In this study, we present analysis of a zebrafish mutant, bugeye, which shows complex inheritance of multiple ocular phenotypes that are known risk factors for glaucoma, including high myopia, elevated intraocular pressure, and up-regulation of stress-response genes in retinal ganglion cells. Molecular genetic analysis revealed that mutations in low density lipoprotein receptor-related protein 2 (lrp2) underlie the mutant phenotypes. Lrp2 is a large transmembrane protein expressed in epithelia of the eye. It facilitates transport and clearance of multiple secreted bioactive factors through receptor-mediated endocytosis. Glaucoma, a progressive blinding disorder, usually presents in adulthood and is characterized by optic nerve damage followed by ganglion cell death. In bugeye/lrp2 mutants, ganglion cell death was significantly elevated, but surprisingly moderate, and therefore they do not model this endpoint of glaucoma. As such, bugeye/lrp2 mutants should be considered valuable as a genetic model (A) for buphthalmia, myopia, and regulated eye growth; (B) for identifying genes and pathways that modify the observed ocular phenotypes; and (C) for studying the initiation of retinal ganglion cell pathology in the context of high myopia and elevated intraocular pressure.
Collapse
Affiliation(s)
- Kerry N. Veth
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Jason R. Willer
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Ross F. Collery
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Matthew P. Gray
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Gregory B. Willer
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Daniel S. Wagner
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, United States of America
| | - Mary C. Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Medical School, Philadelphia, Pennsylvania, United States of America
| | - Ava J. Udvadia
- Department of Biological Sciences, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin, United States of America
| | - Richard S. Smith
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Simon W. M. John
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Ronald G. Gregg
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Brian A. Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
32
|
Kusik BW, Hammond DR, Udvadia AJ. Transcriptional regulatory regions of gap43 needed in developing and regenerating retinal ganglion cells. Dev Dyn 2010; 239:482-95. [PMID: 20034105 DOI: 10.1002/dvdy.22190] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mammals and fish differ in their ability to express axon growth-associated genes in response to CNS injury, which contributes to the differences in their ability for CNS regeneration. Previously we demonstrated that for the axon growth-associated gene, gap43, regions of the rat promoter that are sufficient to promote reporter gene expression in the developing zebrafish nervous system are not sufficient to promote expression in regenerating retinal ganglion cells in zebrafish. Recently, we identified a 3.6-kb gap43 promoter fragment from the pufferfish, Takifugu rubripes (fugu), that can promote reporter gene expression during both development and regeneration. Using promoter deletion analysis, we have found regions of the 3.6-kb fugu gap43 promoter that are necessary for expression in regenerating, but not developing, retinal ganglion cells. Within the 3.6-kb promoter, we have identified elements that are highly conserved among fish, as well as elements conserved among fish, mammals, and birds.
Collapse
Affiliation(s)
- Brandon W Kusik
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | | | | |
Collapse
|
33
|
Fan CY, Cowden J, Simmons SO, Padilla S, Ramabhadran R. Gene expression changes in developing zebrafish as potential markers for rapid developmental neurotoxicity screening. Neurotoxicol Teratol 2010; 32:91-8. [DOI: 10.1016/j.ntt.2009.04.065] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2008] [Revised: 03/25/2009] [Accepted: 04/23/2009] [Indexed: 01/23/2023]
|
34
|
Schmoldt A, Forecki J, Hammond DR, Udvadia AJ. Exploring differential gene expression in zebrafish to teach basic molecular biology skills. Zebrafish 2009; 6:187-99. [PMID: 19250030 DOI: 10.1089/zeb.2008.0574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In an effort to engage students in original research while teaching them basic molecular biology skills, we have designed a course for upper level undergraduate students and beginning graduate students that employs in situ hybridization in whole-mount zebrafish embryos to explore the concept of differential gene regulation. The course was taught in a workshop format during a break between the normal fall and spring semesters, which allowed students to immerse themselves in the concepts and techniques full time over a 13-day period. Overall, the course was successful in exposing students to a variety of techniques in the context of an ongoing research project in our laboratory, which provided beneficial outcomes for students and instructors alike. Here we provide a detailed account of the course organization and preparation, as well as an analysis of learning outcomes achieved by the students.
Collapse
Affiliation(s)
- Angela Schmoldt
- Department of Biological Sciences, University of Wisconsin-Milwaukee, 600 E. Greenfield Ave., Milwaukee, WI 53204, USA
| | | | | | | |
Collapse
|
35
|
Servili A, Bufalino MR, Nishikawa R, de Melo IS, Muñoz-Cueto JA, Lee LE. Establishment of long term cultures of neural stem cells from adult sea bass, Dicentrarchus labrax. Comp Biochem Physiol A Mol Integr Physiol 2009; 152:245-54. [DOI: 10.1016/j.cbpa.2008.10.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 10/10/2008] [Accepted: 10/14/2008] [Indexed: 12/31/2022]
|
36
|
Recent Papers on Zebrafish and Other Aquarium Fish Models. Zebrafish 2008. [DOI: 10.1089/zeb.2008.9987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|