Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Zebrafish ; 21(2): 128-136, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38621212

ABSTRACT

Coordinated signaling pathway activity directs early patterning to set up the vertebrate body plan. Perturbations in the timing or location of signal molecule expression impacts embryo morphology and organ formation. In this study, we present a laboratory course to use zebrafish for studying the role of Wnt signaling in specifying the early embryonic axes. Students are exposed to basic techniques in molecular and developmental biology, including embryo manipulation, fluorescence microscopy, image processing, and data analysis. Furthermore, this course incorporates student-designed experiments to stimulate independent inquiry and improve scientific learning, providing an experience resembling graduate-level laboratory research. Students appreciated following vertebrate development in real-time, and principles of embryogenesis were reinforced by observing the morphological changes that arise due to signaling alterations. Scientific and research skills were enhanced through practice in experimental design, interpretation, and presentation.


Subject(s)
Wnt Signaling Pathway , Zebrafish , Humans , Animals , Zebrafish/genetics , Body Patterning , Embryonic Development , Zebrafish Proteins/metabolism , Gene Expression Regulation, Developmental , Embryo, Nonmammalian/metabolism
2.
FEBS Lett ; 597(2): 262-275, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36217213

ABSTRACT

Mitochondria, organelles critical for energy production, modify their shape and location in response to developmental state and metabolic demands. Mitochondria are altered in diabetes, but the mechanistic basis is poorly defined, due to difficulties in assessing mitochondria within an intact organism. Here, we use in vivo imaging in transparent zebrafish larvae to demonstrate filamentous, interconnected mitochondrial networks within islet cells. Mitochondrial movements highly resemble what has been reported for human islet cells in vitro, showing conservation in behaviour across species and cellular context. During islet development, mitochondrial content increases with emergence of cell motility, and mitochondria disperse within fine protrusions. Overall, this work presents quantitative analysis of mitochondria within their native environment and provides insights into mitochondrial behaviour during organogenesis.


Subject(s)
Mitochondria , Zebrafish , Animals , Humans , Zebrafish/metabolism , Larva , Mitochondria/metabolism , Morphogenesis , Cell Movement
3.
Cells ; 10(11)2021 11 22.
Article in English | MEDLINE | ID: mdl-34831487

ABSTRACT

Diabetic retinopathy is a frequent complication of longstanding diabetes, which comprises a complex interplay of microvascular abnormalities and neurodegeneration. Zebrafish harboring a homozygous mutation in the pancreatic transcription factor pdx1 display a diabetic phenotype with survival into adulthood, and are therefore uniquely suitable among zebrafish models for studying pathologies associated with persistent diabetic conditions. We have previously shown that, starting at three months of age, pdx1 mutants exhibit not only vascular but also neuro-retinal pathologies manifesting as photoreceptor dysfunction and loss, similar to human diabetic retinopathy. Here, we further characterize injury and regenerative responses and examine the effects on progenitor cell populations. Consistent with a negative impact of hyperglycemia on neurogenesis, stem cells of the ciliary marginal zone show an exacerbation of aging-related proliferative decline. In contrast to the robust Müller glial cell proliferation seen following acute retinal injury, the pdx1 mutant shows replenishment of both rod and cone photoreceptors from slow-cycling, neurod-expressing progenitors which first accumulate in the inner nuclear layer. Overall, we demonstrate a diabetic retinopathy model which shows pathological features of the human disease evolving alongside an ongoing restorative process that replaces lost photoreceptors, at the same time suggesting an unappreciated phenotypic continuum between multipotent and photoreceptor-committed progenitors.


Subject(s)
Hyperglycemia/pathology , Neural Stem Cells/pathology , Retina/pathology , Aging/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Death , Cell Proliferation , Chronic Disease , Ependymoglial Cells/pathology , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Models, Biological , Mutation/genetics , Nerve Tissue Proteins/metabolism , PAX6 Transcription Factor/metabolism , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Receptors, Notch/metabolism , Retina/immunology , Signal Transduction , Trans-Activators/genetics , Zebrafish
4.
Front Cell Dev Biol ; 8: 586651, 2020.
Article in English | MEDLINE | ID: mdl-33102488

ABSTRACT

Pancreatic islets, discrete microorgans embedded within the exocrine pancreas, contain beta cells which are critical for glucose homeostasis. Loss or dysfunction of beta cells leads to diabetes, a disease with expanding global prevalence, and for which regenerative therapies are actively being pursued. Recent efforts have focused on producing mature beta cells in vitro, but it is increasingly recognized that achieving a faithful three-dimensional islet structure is crucial for generating fully functional beta cells. Our current understanding of islet morphogenesis is far from complete, due to the deep internal location of the pancreas in mammalian models, which hampers direct visualization. Zebrafish is a model system well suited for studies of pancreas morphogenesis due to its transparency and the accessible location of the larval pancreas. In order to further clarify the cellular mechanisms of islet formation, we have developed new tools for in vivo visualization of single-cell dynamics. Our results show that clustering islet cells make contact and interconnect through dynamic actin-rich processes, move together while remaining in close proximity to the duct, and maintain high protrusive motility after forming clusters. Quantitative analyses of cell morphology and motility in 3-dimensions lays the groundwork to define therapeutically applicable factors responsible for orchestrating the morphogenic behaviors of coalescing endocrine cells.

5.
Invest Ophthalmol Vis Sci ; 61(2): 43, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32106290

ABSTRACT

Purpose: Diabetic retinopathy (DR) is a leading cause of vision impairment and blindness worldwide in the working-age population, and the incidence is rising. Until now it has been difficult to define initiating events and disease progression at the molecular level, as available diabetic rodent models do not present the full spectrum of neural and vascular pathologies. Zebrafish harboring a homozygous mutation in the pancreatic transcription factor pdx1 were previously shown to display a diabetic phenotype from larval stages through adulthood. In this study, pdx1 mutants were examined for retinal vascular and neuronal pathology to demonstrate suitability of these fish for modeling DR. Methods: Vessel morphology was examined in pdx1 mutant and control fish expressing the fli1a:EGFP transgene. We further characterized vascular and retinal phenotypes in mutants and controls using immunohistochemistry, histology, and electron microscopy. Retinal function was assessed using electroretinography. Results: Pdx1 mutants exhibit clear vascular phenotypes at 2 months of age, and disease progression, including arterial vasculopenia, capillary tortuosity, and hypersprouting, could be detected at stages extending over more than 1 year. Neural-retinal pathologies are consistent with photoreceptor dysfunction and loss, but do not progress to blindness. Conclusions: This study highlights pdx1 mutant zebrafish as a valuable complement to rodent and other mammalian models of DR, in particular for research into the mechanistic interplay of diabetes with vascular and neuroretinal disease. They are furthermore suited for molecular studies to identify new targets for treatment of early as well as late DR.


Subject(s)
Diabetic Retinopathy/pathology , Photoreceptor Cells/pathology , Retinal Degeneration/pathology , Retinal Vessels/pathology , Analysis of Variance , Animals , Diabetes Mellitus, Experimental , Diabetic Retinopathy/physiopathology , Electroretinography , Retinal Degeneration/physiopathology , Retinal Vessels/physiopathology , Zebrafish
6.
BMC Biol ; 17(1): 61, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31362746

ABSTRACT

BACKGROUND: FoxH1 is a forkhead transcription factor with conserved key functions in vertebrate mesoderm induction and left-right patterning downstream of the TGF-beta/Nodal signaling pathway. Binding of the forkhead domain (FHD) of FoxH1 to a highly conserved proximal sequence motif was shown to regulate target gene expression. RESULTS: We identify the conserved microRNA-430 family (miR-430) as a novel target of FoxH1. miR-430 levels are increased in foxH1 mutants, resulting in a reduced expression of transcripts that are targeted by miR-430 for degradation. To determine the underlying mechanism of miR-430 repression, we performed chromatin immunoprecipitation studies and overexpression experiments with mutant as well as constitutive active and repressive forms of FoxH1. Our studies reveal a molecular interaction of FoxH1 with miR-430 loci independent of the FHD. Furthermore, we show that previously described mutant forms of FoxH1 that disrupt DNA binding or that lack the C-terminal Smad Interaction Domain (SID) dominantly interfere with miR-430 repression, but not with the regulation of previously described FoxH1 targets. CONCLUSIONS: We were able to identify the distinct roles of protein domains of FoxH1 in the regulation process of miR-430. We provide evidence that the indirect repression of miR-430 loci depends on the connection to a distal repressive chromosome environment via a non-canonical mode. The widespread distribution of such non-canonical binding sites of FoxH1, found not only in our study, argues against a function restricted to regulating miR-430 and for a more global role of FoxH1 in chromatin folding.


Subject(s)
Embryonic Development/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , Zebrafish/genetics , Animals , Embryo, Nonmammalian/metabolism , Forkhead Transcription Factors/metabolism , MicroRNAs/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Development ; 145(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29386244

ABSTRACT

The three-dimensional architecture of the pancreatic islet is integral to beta cell function, but the process of islet formation remains poorly understood due to the difficulties of imaging internal organs with cellular resolution. Within transparent zebrafish larvae, the developing pancreas is relatively superficial and thus amenable to live imaging approaches. We performed in vivo time-lapse and longitudinal imaging studies to follow islet development, visualizing both naturally occurring islet cells and cells arising with an accelerated timecourse following an induction approach. These studies revealed previously unappreciated fine dynamic protrusions projecting between neighboring and distant endocrine cells. Using pharmacological compound and toxin interference approaches, and single-cell analysis of morphology and cell dynamics, we determined that endocrine cell motility is regulated by phosphoinositide 3-kinase (PI3K) and G-protein-coupled receptor (GPCR) signaling. Linking cell dynamics to islet formation, perturbation of protrusion formation disrupted endocrine cell coalescence, and correlated with decreased islet cell differentiation. These studies identified novel cell behaviors contributing to islet morphogenesis, and suggest a model in which dynamic exploratory filopodia establish cell-cell contacts that subsequently promote cell clustering.


Subject(s)
Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Zebrafish/metabolism , Actin Cytoskeleton/metabolism , Animals , Animals, Genetically Modified , Cell Aggregation , Cell Movement , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Islets of Langerhans/cytology , Keratin-18/genetics , Keratin-18/metabolism , Organogenesis , Phosphoinositide-3 Kinase Inhibitors , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Single-Cell Analysis , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
8.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916275

ABSTRACT

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Subject(s)
Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
9.
Sci Rep ; 5: 14241, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26384018

ABSTRACT

Diabetes mellitus is characterized by disrupted glucose homeostasis due to loss or dysfunction of insulin-producing beta cells. In this work, we characterize pancreatic islet development and function in zebrafish mutant for pdx1, a gene which in humans is linked to genetic forms of diabetes and is associated with increased susceptibility to Type 2 diabetes. Pdx1 mutant zebrafish have the key diabetic features of reduced beta cells, decreased insulin and elevated glucose. The hyperglycemia responds to pharmacologic anti-diabetic treatment and, as often seen in mammalian diabetes models, beta cells of pdx1 mutants show sensitivity to nutrient overload. This unique genetic model of diabetes provides a new tool for elucidating the mechanisms behind hyperglycemic pathologies and will allow the testing of novel therapeutic interventions in a model organism that is amenable to high-throughput approaches.


Subject(s)
Animal Feed , Animal Nutritional Physiological Phenomena , Homeodomain Proteins/genetics , Hypoglycemic Agents/pharmacology , Mutation , Trans-Activators/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Body Size , Cell Survival/genetics , Codon, Nonsense , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Gene Knockout Techniques , Genotype , Glucose/metabolism , Homeodomain Proteins/chemistry , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Molecular Sequence Data , Sequence Alignment , Trans-Activators/chemistry , Zebrafish
10.
BMC Biol ; 9: 75, 2011 Oct 31.
Article in English | MEDLINE | ID: mdl-22034951

ABSTRACT

BACKGROUND: Insulin-producing beta cells emerge during pancreas development in two sequential waves. Recently described later-forming beta cells in zebrafish show high similarity to second wave mammalian beta cells in developmental capacity. Loss-of-function studies in mouse and zebrafish demonstrated that the homeobox transcription factors Pdx1 and Hb9 are both critical for pancreas and beta cell development and discrete stage-specific requirements for these genes have been uncovered. Previously, exocrine and endocrine cell recovery was shown to follow loss of pdx1 in zebrafish, but the progenitor cells and molecular mechanisms responsible have not been clearly defined. In addition, interactions of pdx1 and hb9 in beta cell formation have not been addressed. RESULTS: To learn more about endocrine progenitor specification, we examined beta cell formation following morpholino-mediated depletion of pdx1 and hb9. We find that after early beta cell reduction, recovery occurs following loss of either pdx1 or hb9 function. Unexpectedly, simultaneous knockdown of both hb9 and pdx1 leads to virtually complete and persistent beta cell deficiency. We used a NeuroD:EGFP transgenic line to examine endocrine cell behavior in vivo and developed a novel live-imaging technique to document emergence and migration of late-forming endocrine precursors in real time. Our data show that Notch-responsive progenitors for late-arising endocrine cells are predominantly post mitotic and depend on pdx1. By contrast, early-arising endocrine cells are specified and differentiate independent of pdx1. CONCLUSIONS: The nearly complete beta cell deficiency after combined loss of hb9 and pdx1 suggests functional cooperation, which we clarify as distinct roles in early and late endocrine cell formation. A novel imaging approach permitted visualization of the emergence of late endocrine cells within developing embryos for the first time. We demonstrate a pdx1-dependent progenitor population essential for the formation of duct-associated, second wave endocrine cells. We further reveal an unexpectedly low mitotic activity in these progenitor cells, indicating that they are set aside early in development.


Subject(s)
Body Patterning , Endocrine System/embryology , Endocrine System/pathology , Homeodomain Proteins/metabolism , Stem Cells/pathology , Trans-Activators/metabolism , Zebrafish/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Endocrine System/drug effects , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mitosis/drug effects , Models, Biological , Morpholinos/pharmacology , Nerve Tissue Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
11.
Methods Cell Biol ; 100: 261-80, 2010.
Article in English | MEDLINE | ID: mdl-21111221

ABSTRACT

The pancreas is a vertebrate-specific organ of endodermal origin which is responsible for production of digestive enzymes and hormones involved in regulating glucose homeostasis, in particular insulin, deficiency of which results in diabetes. Basic research on the genetic and molecular pathways regulating pancreas formation and function has gained major importance for the development of regenerative medical approaches aimed at improving diabetes treatment. Among the different model organisms that are currently used to elucidate the basic pathways of pancreas development and regeneration, the zebrafish is distinguished by its unique opportunities to combine genetic and pharmacological approaches with sophisticated live-imaging methodology, and by its ability to regenerate the pancreas within a short time. Here we review current perspectives and present methods for studying two important processes contributing to pancreas development and regeneration, namely cell migration via time-lapse micropscopy and cell proliferation via incorporation of nucleotide analog EdU, with a focus on the insulin-producing beta cells of the islet.


Subject(s)
Pancreas/embryology , Pancreas/physiology , Zebrafish/embryology , Animals , Cell Proliferation , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Pancreas/cytology , Zebrafish/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...