Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Mol Cell ; 71(2): 216-228.e7, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30029002

ABSTRACT

The polyglutamine (polyQ) diseases are a group of nine neurodegenerative diseases caused by the expansion of a polyQ tract that results in protein aggregation. Unlike other model organisms, Dictyostelium discoideum is a proteostatic outlier, naturally encoding long polyQ tracts yet resistant to polyQ aggregation. Here we identify serine-rich chaperone protein 1 (SRCP1) as a molecular chaperone that is necessary and sufficient to suppress polyQ aggregation. SRCP1 inhibits aggregation of polyQ-expanded proteins, allowing for their degradation via the proteasome, where SRCP1 is also degraded. SRCP1's C-terminal domain is essential for its activity in cells, and peptides that mimic this domain suppress polyQ aggregation in vitro. Together our results identify a novel type of molecular chaperone and reveal how nature has dealt with the problem of polyQ aggregation.


Subject(s)
Molecular Chaperones/metabolism , Peptides/metabolism , Cell Line , Dictyostelium/metabolism , HEK293 Cells , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Serine/metabolism , Ubiquitin/metabolism
2.
Arterioscler Thromb Vasc Biol ; 43(7): e231-e237, 2023 07.
Article in English | MEDLINE | ID: mdl-37128914

ABSTRACT

BACKGROUND: The goal of this study was to identify and characterize cell-cell interactions that facilitate endothelial tip cell fusion downstream of BMP (bone morphogenic protein)-mediated venous plexus formation. METHODS: High resolution and time-lapse imaging of transgenic reporter lines and loss-of-function studies were carried out to study the involvement of mesenchymal stromal cells during venous angiogenesis. RESULTS: BMP-responsive stromal cells facilitate timely and precise fusion of venous tip cells during developmental angiogenesis. CONCLUSIONS: Stromal cells are required for anastomosis of venous tip cells in the embryonic caudal hematopoietic tissue.


Subject(s)
Bone Morphogenetic Proteins , Mesenchymal Stem Cells , Animals , Cell Fusion , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Animals, Genetically Modified , Cell Communication , Stromal Cells/metabolism
3.
Cell ; 138(6): 1195-208, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19766571

ABSTRACT

Progenitor cell nuclei in the rapidly expanding epithelium of the embryonic vertebrate central nervous system undergo a process called interkinetic nuclear migration (IKNM). Movements of IKNM are generally believed to involve smooth migration of nuclei from apical to basal and back during the G1 and G2 phases of the cell cycle, respectively. Yet, this has not been formally demonstrated, nor have the molecular mechanisms that drive IKNM been identified. Using time-lapse confocal microscopy to observe nuclear movements in zebrafish retinal neuroepithelial cells, we show that, except for brief apical nuclear translocations preceding mitosis, IKNM is stochastic rather than smooth and directed. We also show that IKNM is driven largely by actomyosin-dependent forces as it still occurs when the microtubule cytoskeleton is compromised but is blocked when MyosinII activity is inhibited.


Subject(s)
Actomyosin/metabolism , Cell Nucleus/metabolism , Retina/cytology , Zebrafish/embryology , Animals , Dynactin Complex , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Microtubule-Associated Proteins/metabolism , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Retina/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
4.
Development ; 147(12)2020 06 22.
Article in English | MEDLINE | ID: mdl-32439761

ABSTRACT

The development of the biliary system is a complex yet poorly understood process, with relevance to multiple diseases, including biliary atresia, choledochal cysts and gallbladder agenesis. We present here a crucial role for Hippo-Yap/Taz signaling in this context. Analysis of sav1 mutant zebrafish revealed dysplastic morphology and expansion of both intrahepatic and extrahepatic biliary cells, and ultimately larval lethality. Biliary dysgenesis, but not larval lethality, is driven primarily by Yap signaling. Re-expression of Sav1 protein in sav1-/- hepatocytes is able to overcome these initial deficits and allows sav1-/- fish to survive, suggesting cell non-autonomous signaling from hepatocytes. Examination of sav1-/- rescued adults reveals loss of gallbladder and formation of dysplastic cell masses expressing biliary markers, suggesting roles for Hippo signaling in extrahepatic biliary carcinomas. Deletion of stk3 revealed that the phenotypes observed in sav1 mutant fish function primarily through canonical Hippo signaling and supports a role for phosphatase PP2A, but also suggests Sav1 has functions in addition to facilitating Stk3 activity. Overall, this study defines a role for Hippo-Yap signaling in the maintenance of both intra- and extrahepatic biliary ducts.


Subject(s)
Biliary Tract/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Acyltransferases , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Biliary Tract/anatomy & histology , Biliary Tract/growth & development , CRISPR-Cas Systems/genetics , Carboxylic Ester Hydrolases/metabolism , Gallbladder/anatomy & histology , Gallbladder/growth & development , Gallbladder/metabolism , Larva/growth & development , Larva/metabolism , Liver/anatomy & histology , Liver/metabolism , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Serine-Threonine Kinase 3 , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , YAP-Signaling Proteins , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
5.
Development ; 147(16)2020 08 25.
Article in English | MEDLINE | ID: mdl-32843528

ABSTRACT

The Hippo-Yap pathway regulates multiple cellular processes in response to mechanical and other stimuli. In Drosophila, the polarity protein Lethal (2) giant larvae [L(2)gl], negatively regulates Hippo-mediated transcriptional output. However, in vertebrates, little is known about its homolog Llgl1. Here, we define a novel role for vertebrate Llgl1 in regulating Yap stability in cardiomyocytes, which impacts heart development. In contrast to the role of Drosophila L(2)gl, Llgl1 depletion in cultured rat cardiomyocytes decreased Yap protein levels and blunted target gene transcription without affecting Yap transcript abundance. Llgl1 depletion in zebrafish resulted in larger and dysmorphic cardiomyocytes, pericardial effusion, impaired blood flow and aberrant valvulogenesis. Cardiomyocyte Yap protein levels were decreased in llgl1 morphants, whereas Notch, which is regulated by hemodynamic forces and participates in valvulogenesis, was more broadly activated. Consistent with the role of Llgl1 in regulating Yap stability, cardiomyocyte-specific overexpression of Yap in Llgl1-depleted embryos ameliorated pericardial effusion and restored blood flow velocity. Altogether, our data reveal that vertebrate Llgl1 is crucial for Yap stability in cardiomyocytes and its absence impairs cardiac development.


Subject(s)
Cell Cycle Proteins/metabolism , Heart/embryology , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Cycle Proteins/genetics , Protein Stability , Trans-Activators/genetics , YAP-Signaling Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Development ; 147(22)2020 11 19.
Article in English | MEDLINE | ID: mdl-33060129

ABSTRACT

Adherens junction remodeling regulated by apical polarity proteins constitutes a major driving force for tissue morphogenesis, although the precise mechanism remains inconclusive. Here, we report that, in zebrafish, the Crumbs complex component MPP5a interacts with small GTPase Rab11 in Golgi to transport cadherin and Crumbs components synergistically to the apical domain, thus establishing apical epithelial polarity and adherens junctions. In contrast, Par complex recruited by MPP5a is incapable of interacting with Rab11 but might assemble cytoskeleton to facilitate cadherin exocytosis. In accordance, dysfunction of MPP5a induces an invasive migration of epithelial cells. This adherens junction remodeling pattern is frequently observed in zebrafish lens epithelial cells and neuroepithelial cells. The data identify an unrecognized MPP5a-Rab11 complex and describe its essential role in guiding apical polarization and zonula adherens formation in epithelial cells.


Subject(s)
Adherens Junctions/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Guanylate Cyclase/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , rab GTP-Binding Proteins/metabolism , Adherens Junctions/genetics , Animals , Cadherins/genetics , Cadherins/metabolism , Epithelial Cells , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Guanylate Cyclase/genetics , Protein Transport/physiology , Zebrafish/genetics , Zebrafish Proteins/genetics , rab GTP-Binding Proteins/genetics
7.
Cell ; 134(6): 1055-65, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18805097

ABSTRACT

The different cell types in the central nervous system develop from a common pool of progenitor cells. The nuclei of progenitors move between the apical and basal surfaces of the neuroepithelium in phase with their cell cycle, a process termed interkinetic nuclear migration (INM). In the retina of zebrafish mikre oko (mok) mutants, in which the motor protein Dynactin-1 is disrupted, interkinetic nuclei migrate more rapidly and deeply to the basal side and more slowly to the apical side. We found that Notch signaling is predominantly activated on the apical side in both mutants and wild-type. Mutant progenitors are, thus, less exposed to Notch and exit the cell cycle prematurely. This leads to an overproduction of early-born retinal ganglion cells (RGCs) at the expense of later-born interneurons and glia. Our data indicate that the function of INM is to balance the exposure of progenitor nuclei to neurogenic versus proliferative signals.


Subject(s)
Cell Nucleus/metabolism , Neuroepithelial Cells/cytology , Organogenesis , Retina/embryology , Animals , Body Patterning , Cell Cycle , Cell Differentiation , Dynactin Complex , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Microtubule-Associated Proteins/genetics , Mutation , Neuroepithelial Cells/metabolism , Receptors, Notch/metabolism , Retina/cytology , Retinal Ganglion Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Zebrafish , Zebrafish Proteins/genetics
8.
Semin Cell Dev Biol ; 100: 11-19, 2020 04.
Article in English | MEDLINE | ID: mdl-31606277

ABSTRACT

The response of the adult mammalian heart to injury such as myocardial infarction has long been described as primarily fibrotic scarring and adverse remodeling with little to no regeneration of cardiomyocytes. Emerging studies have challenged this paradigm by demonstrating that, indeed, adult mammalian cardiomyocytes are capable of completing cytokinesis albeit at levels vastly insufficient to compensate for the loss of functional cardiomyocytes following ischemic injury. Thus, there is great interest in identifying mechanisms to guide adult cardiomyocyte cell cycle re-entry and facilitate endogenous heart regeneration. The Hippo signaling pathway is a core kinase cascade that functions to suppress the transcriptional co-activators Yap and Taz by phosphorylation and therefore cytoplasmic retention or phospho-degradation. This pathway has recently sparked interest in the field of cardiac regeneration as inhibition of Hippo kinase signaling or overdriving the transcriptional co-activator, Yap, significantly promotes proliferation of terminally differentiated adult mammalian cardiomyocytes and can restore function in failing mouse hearts. Thus, the Hippo pathway is an attractive therapeutic target for promoting cardiomyocyte renewal and cardiac regeneration. Although the core kinases and transcriptional activators of the Hippo pathway have been studied extensively over the last twenty years, the regulatory inputs of this pathway, particularly in vertebrates, are poorly understood. Recent studies have elucidated several upstream regulatory inputs to the Hippo pathway in adult mammalian cardiomyocytes that influence cell proliferation and heart regeneration. Considering upstream inputs to the Hippo pathway are thought to be context and cell type specific, targeting these various components could serve as a therapeutic approach for refining Hippo-Yap signaling in the heart. Here, we provide an overview of the emerging regulatory inputs to the Hippo pathway as they relate to mammalian cardiomyocytes and heart regeneration.


Subject(s)
Cell Cycle Proteins/metabolism , Heart/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Regeneration , Signal Transduction , Transcription Factors/metabolism , Hippo Signaling Pathway , Humans
9.
PLoS Genet ; 15(1): e1007939, 2019 01.
Article in English | MEDLINE | ID: mdl-30695061

ABSTRACT

The retinal pigment epithelium (RPE) is a specialized monolayer of pigmented cells within the eye that is critical for maintaining visual system function. Diseases affecting the RPE have dire consequences for vision, and the most prevalent of these is atrophic (dry) age-related macular degeneration (AMD), which is thought to result from RPE dysfunction and degeneration. An intriguing possibility for treating RPE degenerative diseases like atrophic AMD is the stimulation of endogenous RPE regeneration; however, very little is known about the mechanisms driving successful RPE regeneration in vivo. Here, we developed a zebrafish transgenic model (rpe65a:nfsB-eGFP) that enabled ablation of large swathes of mature RPE. RPE ablation resulted in rapid RPE degeneration, as well as degeneration of Bruch's membrane and underlying photoreceptors. Using this model, we demonstrate for the first time that zebrafish are capable of regenerating a functional RPE monolayer after RPE ablation. Regenerated RPE cells first appear at the periphery of the RPE, and regeneration proceeds in a peripheral-to-central fashion. RPE ablation elicits a robust proliferative response in the remaining RPE. Subsequently, proliferative cells move into the injury site and differentiate into RPE. BrdU incorporation assays demonstrate that the regenerated RPE is likely derived from remaining peripheral RPE cells. Pharmacological disruption using IWR-1, a Wnt signaling antagonist, significantly reduces cell proliferation in the RPE and impairs overall RPE recovery. These data demonstrate that the zebrafish RPE possesses a robust capacity for regeneration and highlight a potential mechanism through which endogenous RPE regenerate in vivo.


Subject(s)
Macular Degeneration/genetics , Regeneration/genetics , Retinal Pigment Epithelium/growth & development , cis-trans-Isomerases/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Apoptosis/genetics , Bruch Membrane/growth & development , Bruch Membrane/metabolism , Cell Differentiation/genetics , Disease Models, Animal , Green Fluorescent Proteins/genetics , Humans , Imides/administration & dosage , Larva/genetics , Larva/growth & development , Macular Degeneration/pathology , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Quinolines/administration & dosage , Retina/growth & development , Retina/pathology , Retinal Pigment Epithelium/metabolism , Wnt Signaling Pathway/drug effects , Zebrafish/genetics , Zebrafish/growth & development
10.
Development ; 145(9)2018 May 04.
Article in English | MEDLINE | ID: mdl-29678815

ABSTRACT

Balancing the rate of differentiation and proliferation in developing tissues is essential to produce organs of robust size and composition. Although many molecular regulators have been established, how these connect to physical and geometrical aspects of tissue architecture is poorly understood. Here, using high-resolution timelapse imaging, we find that changes to cell geometry associated with dense tissue packing play a significant role in regulating differentiation rate in the zebrafish neural tube. Specifically, progenitors that are displaced away from the apical surface due to crowding, tend to differentiate in a Notch-dependent manner. Using simulations we show that interplay between progenitor density, cell shape and changes in differentiation rate could naturally result in negative-feedback control on progenitor cell number. Given these results, we suggest a model whereby differentiation rate is regulated by density dependent effects on cell geometry to: (1) correct variability in cell number; and (2) balance the rates of proliferation and differentiation over development to 'fill' the available space.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Neural Stem Cells/metabolism , Neural Tube/embryology , Neurogenesis/physiology , Zebrafish/embryology , Animals , Neural Stem Cells/cytology , Neural Tube/cytology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
Nature ; 521(7551): 217-221, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25778702

ABSTRACT

Vertebrates have a unique 3D body shape in which correct tissue and organ shape and alignment are essential for function. For example, vision requires the lens to be centred in the eye cup which must in turn be correctly positioned in the head. Tissue morphogenesis depends on force generation, force transmission through the tissue, and response of tissues and extracellular matrix to force. Although a century ago D'Arcy Thompson postulated that terrestrial animal body shapes are conditioned by gravity, there has been no animal model directly demonstrating how the aforementioned mechano-morphogenetic processes are coordinated to generate a body shape that withstands gravity. Here we report a unique medaka fish (Oryzias latipes) mutant, hirame (hir), which is sensitive to deformation by gravity. hir embryos display a markedly flattened body caused by mutation of YAP, a nuclear executor of Hippo signalling that regulates organ size. We show that actomyosin-mediated tissue tension is reduced in hir embryos, leading to tissue flattening and tissue misalignment, both of which contribute to body flattening. By analysing YAP function in 3D spheroids of human cells, we identify the Rho GTPase activating protein ARHGAP18 as an effector of YAP in controlling tissue tension. Together, these findings reveal a previously unrecognised function of YAP in regulating tissue shape and alignment required for proper 3D body shape. Understanding this morphogenetic function of YAP could facilitate the use of embryonic stem cells to generate complex organs requiring correct alignment of multiple tissues.


Subject(s)
Body Size/genetics , Fish Proteins/metabolism , Morphogenesis/genetics , Oryzias/anatomy & histology , Oryzias/embryology , Actomyosin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Fish Proteins/genetics , GTPase-Activating Proteins/metabolism , Genes, Essential/genetics , Gravitation , Humans , Mutation/genetics , Organ Size/genetics , Oryzias/genetics , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
12.
Hum Mol Genet ; 27(10): 1675-1695, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29506241

ABSTRACT

The PITX2 (paired-like homeodomain 2) gene encodes a bicoid-like homeodomain transcription factor linked with several human disorders. The main associated congenital phenotype is Axenfeld-Rieger syndrome, type 1, an autosomal dominant condition characterized by variable defects in the anterior segment of the eye, an increased risk of glaucoma, craniofacial dysmorphism and dental and umbilical anomalies; in addition to this, one report implicated PITX2 in ring dermoid of the cornea and a few others described cardiac phenotypes. We report three novel PITX2 mutations-c.271C > T, p.(Arg91Trp); c.259T > C, p.(Phe87Leu); and c.356delA, p.(Gln119Argfs*36)-identified in independent families with typical Axenfeld-Rieger syndrome characteristics and some unusual features such as corneal guttata, Wolf-Parkinson-White syndrome, and hyperextensibility. To gain further insight into the diverse roles of PITX2/pitx2 in vertebrate development, we generated various genetic lesions in the pitx2 gene via TALEN-mediated genome editing. Affected homozygous zebrafish demonstrated congenital defects consistent with the range of PITX2-associated human phenotypes: abnormal development of the cornea, iris and iridocorneal angle; corneal dermoids; and craniofacial dysmorphism. In addition, via comparison of pitx2M64* and wild-type embryonic ocular transcriptomes we defined molecular changes associated with pitx2 deficiency, thereby implicating processes potentially underlying disease pathology. This analysis identified numerous affected factors including several members of the Wnt pathway and collagen types I and V gene families. These data further support the link between PITX2 and the WNT pathway and suggest a new role in regulation of collagen gene expression during development.


Subject(s)
Anterior Eye Segment/abnormalities , Eye Abnormalities/genetics , Eye Diseases, Hereditary/genetics , Glaucoma/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Wolff-Parkinson-White Syndrome/genetics , Zebrafish Proteins/genetics , Animals , Anterior Eye Segment/physiopathology , Collagen Type I/genetics , Collagen Type V/genetics , Eye Abnormalities/physiopathology , Eye Diseases, Hereditary/physiopathology , Gene Editing , Gene Expression Regulation, Developmental , Glaucoma/physiopathology , Humans , Mutation , Pedigree , Transcription Factors/deficiency , Wnt Signaling Pathway , Zebrafish/genetics , Zebrafish Proteins/deficiency , Homeobox Protein PITX2
13.
J Am Soc Nephrol ; 29(5): 1525-1535, 2018 05.
Article in English | MEDLINE | ID: mdl-29476007

ABSTRACT

Background Interpreting genetic variants is one of the greatest challenges impeding analysis of rapidly increasing volumes of genomic data from patients. For example, SHROOM3 is an associated risk gene for CKD, yet causative mechanism(s) of SHROOM3 allele(s) are unknown.Methods We used our analytic pipeline that integrates genetic, computational, biochemical, CRISPR/Cas9 editing, molecular, and physiologic data to characterize coding and noncoding variants to study the human SHROOM3 risk locus for CKD.Results We identified a novel SHROOM3 transcriptional start site, which results in a shorter isoform lacking the PDZ domain and is regulated by a common noncoding sequence variant associated with CKD (rs17319721, allele frequency: 0.35). This variant disrupted allele binding to the transcription factor TCF7L2 in podocyte cell nuclear extracts and altered transcription levels of SHROOM3 in cultured cells, potentially through the loss of repressive looping between rs17319721 and the novel start site. Although common variant mechanisms are of high utility, sequencing is beginning to identify rare variants involved in disease; therefore, we used our biophysical tools to analyze an average of 112,849 individual human genome sequences for rare SHROOM3 missense variants, revealing 35 high-effect variants. The high-effect alleles include a coding variant (P1244L) previously associated with CKD (P=0.01, odds ratio=7.95; 95% CI, 1.53 to 41.46) that we find to be present in East Asian individuals at an allele frequency of 0.0027. We determined that P1244L attenuates the interaction of SHROOM3 with 14-3-3, suggesting alterations to the Hippo pathway, a known mediator of CKD.Conclusions These data demonstrate multiple new SHROOM3-dependent genetic/molecular mechanisms that likely affect CKD.


Subject(s)
Microfilament Proteins/genetics , Renal Insufficiency, Chronic/genetics , Alleles , Animals , Cell Nucleus , Gene Frequency , Genetic Loci , HEK293 Cells , Humans , Mice , Mutation, Missense , Podocytes , Protein Isoforms/genetics , Transcription Factor 7-Like 2 Protein/genetics , Transcription, Genetic , Zebrafish
14.
Dev Biol ; 425(2): 176-190, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28341548

ABSTRACT

Zebrafish morphants of osm-3/kif17, a kinesin-2 family member and intraflagellar transport motor, have photoreceptor outer segments that are dramatically reduced in number and size. However, two genetic mutant lines, osm-3/kif17sa0119 and osm-3/kif17sa18340, reportedly lack any observable morphological outer segment defects. In this work, we use TALENs to generate an independent allele, osm-3/kif17mw405, and show that both osm-3/kif17sa0119 and osm-3/kif17mw405 have an outer segment developmental delay in both size and density that is fully recovered by 6 days post-fertilization. Additionally, we use CRISPRs to generate cos2/kif7mw406, a mutation in the kinesin-4 family member cos2/kif7 that has been implicated in controlling ciliary architecture and Hedgehog signaling to test whether it may be functioning redundantly with osm-3/kif17. We show that cos2/kif7mw406 has an outer segment developmental delay similar to the osm-3/kif17 mutants. Using a three-dimensional mathematical model of outer segments, we show that while cos2/kif7mw406 and osm-3/kif17mw405 outer segments are smaller throughout the first 6 days of development, the volumetric rates of outer segment morphogenesis are not different among wild-type, cos2/kif7mw406, and osm-3/kif17mw405 after 60hpf. Instead, our model suggests that cos2/kif7mw406 and osm-3/kif17mw405 impact outer segment morphogenesis through upstream events that that are different for each motor. In the case of cos2/kif7mw406 mutants, we show that early defects in Hedgehog signaling lead to a general, non-photoreceptor-specific delay of retinal neurogenesis, which in turn causes the secondary phenotype of delayed outer segment morphogenesis. In contrast, the osm-3/kif17mw405 outer segment morphogenesis delays are linked specifically to initial disc morphogenesis of photoreceptors rather than an upstream event. Further, we show that osm-3/kif17 mutant mice also exhibit a similarly delayed outer segment development, suggesting a role for osm-3/kif17 in early outer segment development that is conserved across species. In conclusion, we show that both osm-3/kif17 and cos2/kif7 have comparable outer segment developmental delays, although through independent mechanisms.


Subject(s)
Kinesins/metabolism , Morphogenesis , Retinal Photoreceptor Cell Outer Segment/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cilia/drug effects , Cilia/metabolism , Gene Editing , Hedgehog Proteins/metabolism , Mice, Inbred C57BL , Models, Biological , Morphogenesis/drug effects , Morpholinos/pharmacology , Mutation/genetics , Neurogenesis/drug effects , Signal Transduction/drug effects , Temperature , Transcription Activator-Like Effector Nucleases
15.
BMC Cell Biol ; 19(1): 25, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30458707

ABSTRACT

BACKGROUND: KIF17, a kinesin-2 motor that functions in intraflagellar transport, can regulate the onset of photoreceptor outer segment development. However, the function of KIF17 in a mature photoreceptor remains unclear. Additionally, the ciliary localization of KIF17 is regulated by a C-terminal consensus sequence (KRKK) that is immediately adjacent to a conserved residue (mouse S1029/zebrafish S815) previously shown to be phosphorylated by CaMKII. Yet, whether this phosphorylation can regulate the localization, and thus function, of KIF17 in ciliary photoreceptors remains unknown. RESULTS: Using transgenic expression in zebrafish photoreceptors, we show that phospho-mimetic KIF17 has enhanced localization along the cone outer segment. Importantly, expression of phospho-mimetic KIF17 is associated with greatly enhanced turnover of the photoreceptor outer segment through disc shedding in a cell-autonomous manner, while genetic mutants of kif17 in zebrafish and mice have diminished disc shedding. Lastly, cone expression of constitutively active tCaMKII leads to a kif17-dependent increase in disc shedding. CONCLUSIONS: Taken together, our data support a model in which phosphorylation of KIF17 promotes its photoreceptor outer segment localization and disc shedding, a process essential for photoreceptor maintenance and homeostasis. While disc shedding has been predominantly studied in the context of the mechanisms underlying phagocytosis of outer segments by the retinal pigment epithelium, this work implicates photoreceptor-derived signaling in the underlying mechanisms of disc shedding.


Subject(s)
Kinesins/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Cilia/metabolism , Humans , Kinesins/chemistry , Mice, Inbred C57BL , Mutation/genetics , Phagosomes/metabolism , Phagosomes/ultrastructure , Phosphorylation , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/ultrastructure , Zebrafish Proteins/chemistry
16.
Genome Res ; 25(1): 57-65, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25273069

ABSTRACT

Genome-wide association studies (GWAS) identify regions of the genome correlated with disease risk but are restricted in their ability to identify the underlying causative mechanism(s). Thus, GWAS are useful "roadmaps" that require functional analysis to establish the genetic and mechanistic structure of a particular locus. Unfortunately, direct functional testing in humans is limited, demonstrating the need for complementary approaches. Here we used an integrated approach combining zebrafish, rat, and human data to interrogate the function of an established GWAS locus (SHROOM3) lacking prior functional support for chronic kidney disease (CKD). Congenic mapping and sequence analysis in rats suggested Shroom3 was a strong positional candidate gene. Transferring a 6.1-Mb region containing the wild-type Shroom3 gene significantly improved the kidney glomerular function in FHH (fawn-hooded hypertensive) rat. The wild-type Shroom3 allele, but not the FHH Shroom3 allele, rescued glomerular defects induced by knockdown of endogenous shroom3 in zebrafish, suggesting that the FHH Shroom3 allele is defective and likely contributes to renal injury in the FHH rat. We also show for the first time that variants disrupting the actin-binding domain of SHROOM3 may cause podocyte effacement and impairment of the glomerular filtration barrier.


Subject(s)
Glomerular Filtration Barrier/metabolism , Microfilament Proteins/metabolism , Zebrafish Proteins/metabolism , Alleles , Amino Acid Sequence , Animals , Animals, Congenic , Animals, Genetically Modified , Cloning, Molecular , Exons , Female , Genetic Loci , Genetic Variation , Genome-Wide Association Study , Humans , Kidney Diseases/genetics , Male , Microfilament Proteins/genetics , Microscopy, Electron, Transmission , Molecular Sequence Data , Plasmids/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Analysis, DNA , Zebrafish , Zebrafish Proteins/genetics
17.
Development ; 142(17): 3021-32, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26209646

ABSTRACT

The optic vesicle comprises a pool of bi-potential progenitor cells from which the retinal pigment epithelium (RPE) and neural retina fates segregate during ocular morphogenesis. Several transcription factors and signaling pathways have been shown to be important for RPE maintenance and differentiation, but an understanding of the initial fate specification and determination of this ocular cell type is lacking. We show that Yap/Taz-Tead activity is necessary and sufficient for optic vesicle progenitors to adopt RPE identity in zebrafish. A Tead-responsive transgene is expressed within the domain of the optic cup from which RPE arises, and Yap immunoreactivity localizes to the nuclei of prospective RPE cells. yap (yap1) mutants lack a subset of RPE cells and/or exhibit coloboma. Loss of RPE in yap mutants is exacerbated in combination with taz (wwtr1) mutant alleles such that, when Yap and Taz are both absent, optic vesicle progenitor cells completely lose their ability to form RPE. The mechanism of Yap-dependent RPE cell type determination is reliant on both nuclear localization of Yap and interaction with a Tead co-factor. In contrast to loss of Yap and Taz, overexpression of either protein within optic vesicle progenitors leads to ectopic pigmentation in a dosage-dependent manner. Overall, this study identifies Yap and Taz as key early regulators of RPE genesis and provides a mechanistic framework for understanding the congenital ocular defects of Sveinsson's chorioretinal atrophy and congenital retinal coloboma.


Subject(s)
Cell Lineage , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Retinal Pigment Epithelium/cytology , Trans-Activators/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Alleles , Animals , Apoptosis/genetics , Cell Nucleus/metabolism , Cell Proliferation , Coloboma/pathology , Gene Expression Regulation, Developmental , Genes, Reporter , HEK293 Cells , Humans , Morphogenesis/genetics , Mutation , Phenotype , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/transplantation , Signal Transduction/genetics , TEA Domain Transcription Factors , Trans-Activators/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transgenes , Up-Regulation , YAP-Signaling Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
18.
EMBO J ; 32(1): 30-44, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23202854

ABSTRACT

By analysing the cellular and subcellular events that occur in the centre of the developing zebrafish neural rod, we have uncovered a novel mechanism of cell polarisation during lumen formation. Cells from each side of the neural rod interdigitate across the tissue midline. This is necessary for localisation of apical junctional proteins to the region where cells intersect the tissue midline. Cells assemble a mirror-symmetric microtubule cytoskeleton around the tissue midline, which is necessary for the trafficking of proteins required for normal lumen formation, such as partitioning defective 3 and Rab11a to this point. This occurs in advance and is independent of the midline cell division that has been shown to have a powerful role in lumen organisation. To our knowledge, this is the first example of the initiation of apical polarisation part way along the length of a cell, rather than at a cell extremity. Although the midline division is not necessary for apical polarisation, it confers a morphogenetic advantage by efficiently eliminating cellular processes that would otherwise bridge the developing lumen.


Subject(s)
Cell Communication , Microtubules/metabolism , Neural Tube/embryology , Neurulation , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Division , Cell Movement , Cell Polarity , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Green Fluorescent Proteins/chemistry , Luminescent Agents/chemistry , Microtubules/genetics , Mutation , Neural Tube/cytology , Nocodazole/pharmacology , Protein Transport/drug effects , Recombinant Fusion Proteins , Tubulin Modulators/pharmacology , Zebrafish/anatomy & histology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
19.
Vis Neurosci ; 33: E011, 2016 01.
Article in English | MEDLINE | ID: mdl-28177275

ABSTRACT

Zebrafish (Danio rerio) provide many advantages as a model organism for studying ocular disease and development, and there is great interest in the ability to non-invasively assess their photoreceptor mosaic. Despite recent applications of scanning light ophthalmoscopy, fundus photography, and gonioscopy to in vivo imaging of the adult zebrafish eye, current techniques either lack accurate scaling information (limiting quantitative analyses) or require euthanizing the fish (precluding longitudinal analyses). Here we describe improved methods for imaging the adult zebrafish retina using spectral domain optical coherence tomography (OCT). Transgenic fli1:eGFP zebrafish were imaged using the Bioptigen Envisu R2200 broadband source OCT with a 12-mm telecentric probe to measure axial length and a mouse retina probe to acquire retinal volume scans subtending 1.2 × 1.2 mm nominally. En face summed volume projections were generated from the volume scans using custom software that allows the user to create contours tailored to specific retinal layer(s) of interest. Following imaging, the eyes were dissected for ex vivo fluorescence microscopy, and measurements of blood vessel branch points were compared to those made from the en face OCT images to determine the OCT lateral scale as a function of axial length. Using this scaling model, we imaged the photoreceptor layer of five wild-type zebrafish and quantified the density and packing geometry of the UV cone submosaic. Our in vivo cone density measurements agreed with measurements from previously published histology values. The method presented here allows accurate, quantitative assessment of cone structure in vivo and will be useful for longitudinal studies of the zebrafish cone mosaics.


Subject(s)
Retinal Cone Photoreceptor Cells/cytology , Tomography, Optical Coherence , Zebrafish/anatomy & histology , Animals , Animals, Genetically Modified , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence , Zebrafish/genetics
20.
Development ; 139(9): 1599-610, 2012 May.
Article in English | MEDLINE | ID: mdl-22492354

ABSTRACT

To gain insights into the cellular mechanisms of neurogenesis, we analyzed retinal neuroepithelia deficient for Llgl1, a protein implicated in apicobasal cell polarity, asymmetric cell division, cell shape and cell cycle exit. We found that vertebrate retinal neuroepithelia deficient for Llgl1 retained overt apicobasal polarity, but had expanded apical domains. Llgl1 retinal progenitors also had increased Notch activity and reduced rates of neurogenesis. Blocking Notch function by depleting Rbpj restored normal neurogenesis. Experimental expansion of the apical domain, through inhibition of Shroom3, also increased Notch activity and reduced neurogenesis. Significantly, in wild-type retina, neurogenic retinal progenitors had smaller apical domains compared with proliferative neuroepithelia. As nuclear position during interkinetic nuclear migration (IKNM) has been previously linked with cell cycle exit, we analyzed this phenomenon in cells depleted of Llgl1. We found that although IKNM was normal, the relationship between nuclear position and neurogenesis was shifted away from the apical surface, consistent with increased pro-proliferative and/or anti-neurogenic signals associated with the apical domain. These data, in conjunction with other findings, suggest that, in retinal neuroepithelia, the size of the apical domain modulates the strength of polarized signals that influence neurogenesis.


Subject(s)
Cell Cycle Proteins/deficiency , Neuroepithelial Cells/metabolism , Neurogenesis/physiology , Receptors, Notch/metabolism , Retina/cytology , Zebrafish Proteins/deficiency , Zebrafish/physiology , Animals , Bromodeoxyuridine , Cell Cycle Proteins/metabolism , Microfilament Proteins/metabolism , Oligonucleotides/genetics , Time-Lapse Imaging , Zebrafish/metabolism , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL