Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963001

ABSTRACT

Semaphorin6A (Sema6A) is a repulsive guidance molecule that plays many roles in central nervous system, heart, and bone development, as well as immune system responses and cell signaling in cancer. Loss of Sema6A or its receptor PlexinA2 in zebrafish leads to smaller eyes and improper retinal patterning. Here we investigate a potential role for the Sema6A intracellular domain in zebrafish eye development and dissect which phenotypes rely on forward signaling and which rely on reverse signaling. We performed rescue experiments on zebrafish Sema6A morphants with either full-length Sema6A (Sema6A-FL) or Sema6A lacking its intracellular domain (Sema6A-ΔC). We identified that the intracellular domain is not required for eye size and retinal patterning, however it is required for retinal integrity, the number and end feet strength of Müller glia and protecting against retinal cell death. This novel function for the intracellular domain suggests a role for Sema6A reverse signaling in zebrafish eye development.

2.
bioRxiv ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38798399

ABSTRACT

Myosin-binding protein H (MyBP-H) is a component of the vertebrate skeletal muscle sarcomere with sequence and domain homology to myosin-binding protein C (MyBP-C). Whereas skeletal muscle isoforms of MyBP-C (fMyBP-C, sMyBP-C) modulate muscle contractility via interactions with actin thin filaments and myosin motors within the muscle sarcomere "C-zone," MyBP-H has no known function. This is in part due to MyBP-H having limited expression in adult fast-twitch muscle and no known involvement in muscle disease. Quantitative proteomics reported here reveal MyBP-H is highly expressed in prenatal rat fast-twitch muscles and larval zebrafish, suggesting a conserved role in muscle development, and promoting studies to define its function. We take advantage of the genetic control of the zebrafish model and a combination of structural, functional, and biophysical techniques to interrogate the role of MyBP-H. Transgenic, FLAG-tagged MyBP-H or fMyBP-C both localize to the C-zones in larval myofibers, whereas genetic depletion of endogenous MyBP-H or fMyBP-C leads to increased accumulation of the other, suggesting competition for C-zone binding sites. Does MyBP-H modulate contractility from the C-zone? Globular domains critical to MyBP-C's modulatory functions are absent from MyBP-H, suggesting MyBP-H may be functionally silent. However, our results suggest an active role. Small angle x-ray diffraction of intact larval tails revealed MyBP-H contributes to the compression of the myofilament lattice accompanying stretch or contraction, while in vitro motility experiments indicate MyBP-H shares MyBP-C's capacity as a molecular "brake". These results provide new insights and raise questions about the role of the C-zone during muscle development.

3.
FEBS Lett ; 598(3): 302-320, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38058169

ABSTRACT

CRK adaptor proteins are important for signal transduction mechanisms driving cell proliferation and positioning during vertebrate central nervous system development. Zebrafish lacking both CRK family members exhibit small, disorganized retinas with 50% penetrance. The goal of this study was to determine whether another adaptor protein might functionally compensate for the loss of CRK adaptors. Expression patterns in developing zebrafish, and bioinformatic analyses of the motifs recognized by their SH2 and SH3 domains, suggest NCK adaptors are well-positioned to compensate for loss of CRK adaptors. In support of this hypothesis, proteomic analyses found CRK and NCK adaptors share overlapping interacting partners including known regulators of cell adhesion and migration, suggesting their functional intersection in neurodevelopment.


Subject(s)
Proteomics , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction/physiology , src Homology Domains
4.
Dev Dyn ; 251(2): 362-376, 2022 02.
Article in English | MEDLINE | ID: mdl-34268820

ABSTRACT

BACKGROUND: The development of the central nervous system (CNS) requires critical cell signaling molecules to coordinate cell proliferation and migration in order to structure the adult tissue. Chicken tumor virus #10 Regulator of Kinase (CRK) and CRK-like (CRKL) are adaptor proteins with pre-metazoan ancestry and are known to be required for patterning laminated structures downstream of Reelin (RELN), such as the cerebral cortex, cerebellum, and hippocampus. CRK and CRKL also play crucial roles in a variety of other growth factor and extracellular matrix signaling cascades. The neuronal retina is another highly laminated structure within the CNS that is dependent on migration for proper development, but the cell signaling mechanisms behind neuronal positioning in the retina are only partly understood. RESULTS: We find that crk and crkl have largely overlapping expression within the developing zebrafish nervous system. We find that their disruption results in smaller eye size and loss of retinal lamination. CONCLUSIONS: Our data indicate that Crk adaptors are critical for proper development of the zebrafish neural retina in a crk/crkl dose-dependent manner.


Subject(s)
Nuclear Proteins , Zebrafish , Animals , Cell Proliferation , Nuclear Proteins/metabolism , Retina/metabolism , Signal Transduction/physiology , Zebrafish/metabolism
5.
FEBS J ; 288(1): 142-159, 2021 01.
Article in English | MEDLINE | ID: mdl-32543048

ABSTRACT

Charcot-Marie-Tooth disease (CMT) encompasses a set of genetically and clinically heterogeneous neuropathies characterized by length-dependent dysfunction of the peripheral nervous system. Mutations in over 80 diverse genes are associated with CMT, and aminoacyl-tRNA synthetases (ARS) constitute a large gene family implicated in the disease. Despite considerable efforts to elucidate the mechanistic link between ARS mutations and the CMT phenotype, the molecular basis of the pathology is unknown. In this work, we investigated the impact of three CMT-associated substitutions (V155G, Y330C, and R137Q) in the cytoplasmic histidyl-tRNA synthetase (HARS1) on neurite outgrowth and peripheral nervous system development. The model systems for this work included a nerve growth factor-stimulated neurite outgrowth model in rat pheochromocytoma cells (PC12), and a zebrafish line with GFP/red fluorescent protein reporters of sensory and motor neuron development. The expression of CMT-HARS1 mutations led to attenuation of protein synthesis and increased phosphorylation of eIF2α in PC12 cells and was accompanied by impaired neurite and axon outgrowth in both models. Notably, these effects were phenocopied by histidinol, a HARS1 inhibitor, and cycloheximide, a protein synthesis inhibitor. The mutant proteins also formed heterodimers with wild-type HARS1, raising the possibility that CMT-HARS1 mutations cause disease through a dominant-negative mechanism. Overall, these findings support the hypothesis that CMT-HARS1 alleles exert their toxic effect in a neuronal context, and lead to dysregulated protein synthesis. These studies demonstrate the value of zebrafish as a model for studying mutant alleles associated with CMT, and for characterizing the processes that lead to peripheral nervous system dysfunction.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Histidine-tRNA Ligase/genetics , Neuronal Outgrowth/genetics , Neurons/metabolism , Peripheral Nervous System/metabolism , Protein Biosynthesis , Animals , Animals, Genetically Modified , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Cycloheximide/pharmacology , Disease Models, Animal , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Histidine-tRNA Ligase/antagonists & inhibitors , Histidine-tRNA Ligase/metabolism , Histidinol/pharmacology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mutation , Neuronal Outgrowth/drug effects , Neurons/drug effects , Neurons/pathology , PC12 Cells , Peripheral Nervous System/pathology , Protein Multimerization , Rats , Zebrafish , Red Fluorescent Protein
6.
Biophys J ; 119(4): 806-820, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32755560

ABSTRACT

Zebrafish (Danio rerio) swim within days of fertilization, powered by muscles of the axial myotomes. Forces generated by these muscles can be measured rapidly in whole, intact larval tails by adapting protocols developed for ex vivo muscle mechanics. But it is not known how well these measurements reflect the function of the underlying muscle fibers and sarcomeres. Here, we consider the anatomy of the 5-day-old, wild-type larval tail, and implement technical modifications to measuring muscle physiology in intact tails. Specifically, we quantify fundamental relationships between force, length, and shortening velocity, and capture the extreme contractile speeds required to swim with tail-beat frequencies of 80-100 Hz. Therefore, we analyze 1000 frames/s videos to track the movement of structures, visible in the transparent tail, which correlate with sarcomere length. We also characterize the passive viscoelastic properties of the preparation to isolate forces contributed by nonmuscle structures within the tail. Myotomal muscles generate more than 95% of their maximal isometric stress (76 ± 3 mN/mm2) over the range of muscle lengths used in vivo. They have rapid twitch kinetics (full width at half-maximal stress: 11 ± 1 ms) and a high twitch/tetanus ratio (0.91 ± 0.05), indicating adaptations for fast excitation-contraction coupling. Although contractile stress is relatively low, myotomal muscles develop high net power (134 ± 20 W/kg at 80 Hz) in cyclical work loop experiments designed to simulate the in vivo dynamics of muscle fibers during swimming. When shortening at a constant speed of 7 ± 1 muscle lengths/s, muscles develop 86 ± 2% of isometric stress, whereas peak instantaneous power during 100 Hz work loops occurs at 18 ± 2 muscle lengths/s. These approaches can improve the usefulness of zebrafish as a model system for muscle research by providing a rapid and sensitive functional readout for experimental interventions.


Subject(s)
Swimming , Zebrafish , Animals , Larva , Muscle Contraction , Sarcomeres
7.
Mol Cell Proteomics ; 19(10): 1586-1601, 2020 10.
Article in English | MEDLINE | ID: mdl-32606017

ABSTRACT

The Discoidin, CUB, and LCCL domain-containing protein (DCBLD) family consists of two type-I transmembrane scaffolding receptors, DCBLD1 and DCBLD2, which play important roles in development and cancer. The nonreceptor tyrosine kinases FYN and ABL are known to drive phosphorylation of tyrosine residues in YXXP motifs within the intracellular domains of DCBLD family members, which leads to the recruitment of the Src homology 2 (SH2) domain of the adaptors CT10 regulator of kinase (CRK) and CRK-like (CRKL). We previously characterized the FYN- and ABL-driven phosphorylation of DCBLD family YXXP motifs. However, we have identified additional FYN- and ABL-dependent phosphorylation sites on DCBLD1 and DCBLD2. This suggests that beyond CRK and CRKL, additional DCBLD interactors may be regulated by FYN and ABL activity. Here, we report a quantitative proteomics approach in which we map the FYN- and ABL-regulated interactomes of DCBLD family members. We found FYN and ABL regulated the binding of several signaling molecules to DCBLD1 and DCBLD2, including members of the 14-3-3 family of adaptors. Biochemical investigation of the DCBLD2/14-3-3 interaction revealed ABL-induced binding of 14-3-3 family members directly to DCBLD2.


Subject(s)
Membrane Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Amino Acid Sequence , HEK293 Cells , Humans , Membrane Proteins/chemistry , Models, Biological , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Phosphorylation , Protein Interaction Maps
8.
Dev Dyn ; 249(10): 1285-1295, 2020 10.
Article in English | MEDLINE | ID: mdl-32406957

ABSTRACT

BACKGROUND: Semaphorin6A (Sema6A) and its PlexinA2 (PlxnA2) receptor canonically function as repulsive axon guidance cues. To understand downstream signaling mechanisms, we performed a microarray screen and identified the "clutch molecule" shootin-1 (shtn-1) as a transcriptionally repressed target. Shtn-1 is a key proponent of cell migration and neuronal polarization and must be regulated during nervous system development. The mechanisms of Shtn-1 regulation and the phenotypic consequences of loss of repression are poorly understood. RESULTS: We demonstrate shtn-1 overexpression results in impaired migration of the optic vesicles, lack of retinal pigmented epithelium, and pathfinding errors of retinotectal projections. We also observed patterning defects in the peripheral nervous system. Importantly, these phenotypes were rescued by overexpressing PlxnA2. CONCLUSIONS: We demonstrate a functional role for repression of shtn-1 by PlxnA2 in development of the eyes and peripheral nervous system in zebrafish. These results demonstrate that careful regulation of shtn-1 is critical for development of the nervous system.


Subject(s)
Cytoskeletal Proteins/physiology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/physiology , Nervous System/embryology , Receptors, Cell Surface/physiology , Semaphorins/physiology , Zebrafish Proteins/physiology , Animals , Axons/physiology , Body Patterning , Cell Movement , Cytoskeletal Proteins/genetics , Humans , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Peripheral Nervous System/physiology , Phenotype , Receptors, Cell Surface/genetics , Retinal Pigment Epithelium/physiology , Semaphorins/genetics , Zebrafish , Zebrafish Proteins/genetics
9.
FEBS Lett ; 593(21): 3015-3028, 2019 11.
Article in English | MEDLINE | ID: mdl-31378926

ABSTRACT

Semaphorins (Semas) are a family of secreted and transmembrane proteins that play critical roles in development. Interestingly, several vertebrate transmembrane Sema classes are capable of producing functional soluble ectodomains. However, little is known of soluble Sema6 ectodomains in the nervous system. Herein, we show that the soluble Sema6A ectodomain, sSema6A, exhibits natural and protein kinase C (PKC)-induced release. We show that PKC mediates Sema6A phosphorylation at specific sites and while this phosphorylation is not the primary mechanism regulating sSema6A production, we found that the intracellular domain confers resistance to ectodomain release. Finally, sSema6A is functional as it promotes the cohesion of zebrafish early eye field explants. This suggests that in addition to its canonical contact-mediated functions, Sema6A may have regulated, long-range, forward-signaling capacity.


Subject(s)
Frontal Lobe/metabolism , Protein Kinase C/metabolism , Semaphorins/chemistry , Semaphorins/metabolism , Zebrafish/growth & development , Animals , Frontal Lobe/cytology , Gene Expression Regulation , HEK293 Cells , Humans , Mass Spectrometry , Mice , Phosphorylation , Protein Domains , Semaphorins/genetics , Serine/chemistry , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Biochem J ; 476(6): 931-950, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30902898

ABSTRACT

The discoidin, CUB, and LCCL domain-containing (DCBLD) receptor family are composed of the type-I transmembrane proteins DCBLD1 and DCBLD2 (also ESDN and CLCP1). These proteins are highly conserved across vertebrates and possess similar domain structure to that of neuropilins, which act as critical co-receptors in developmental processes. Although DCBLD1 remains largely uncharacterized, the functional and mechanistic roles of DCBLD2 are emerging. This review provides a comprehensive discussion of this presumed receptor family, ranging from structural and signaling aspects to their associations with cancer, physiology, and development.


Subject(s)
Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Humans , Neoplasms/pathology
11.
Gene Expr Patterns ; 31: 1-6, 2019 01.
Article in English | MEDLINE | ID: mdl-30468770

ABSTRACT

Protein kinase A (PKA), also known as cAMP dependent protein kinase, is an essential component of many signaling pathways, many of which regulate key developmental processes. Inactive PKA is a tetrameric holoenzyme, comprised of two catalytic (PRKAC), and two regulatory subunits. Upon cAMP binding, the catalytic subunits are released and thereby activated. There are multiple isoforms of PKA catalytic subunits, but their individual roles are not well understood. In order to begin studying their roles in zebrafish development, it is first necessary to identify the spatial and temporal expression profiles for each prkac subunit. Here we evaluate the expression profiles for the four zebrafish prkacs: prkacαa, αb, ßa, and ßb, at key developmental time points: 24, 48 and 72 h post fertilization. We show that zebrafish prkacs are expressed throughout the developing nervous system, each showing unique expression patterns. This body of work will inform future functional studies into the roles of PKA during development.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation, Developmental , Zebrafish Proteins/genetics , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Nervous System/embryology , Nervous System/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Zebrafish , Zebrafish Proteins/metabolism
12.
Bioinformatics ; 34(22): 3898-3906, 2018 11 15.
Article in English | MEDLINE | ID: mdl-29868839

ABSTRACT

Motivation: The development of proteomic methods for the characterization of domain/motif interactions has greatly expanded our understanding of signal transduction. However, proteomics-based binding screens have limitations including that the queried tissue or cell type may not harbor all potential interacting partners or post-translational modifications (PTMs) required for the interaction. Therefore, we sought a generalizable, complementary in silico approach to identify potentially novel motif and PTM-dependent binding partners of high priority. Results: We used as an initial example the interaction between the Src homology 2 (SH2) domains of the adaptor proteins CT10 regulator of kinase (CRK) and CRK-like (CRKL) and phosphorylated-YXXP motifs. Employing well-curated, publicly-available resources, we scored and prioritized potential CRK/CRKL-SH2 interactors possessing signature characteristics of known interacting partners. Our approach gave high priority scores to 102 of the >9000 YXXP motif-containing proteins. Within this 102 were 21 of the 25 curated CRK/CRKL-SH2-binding partners showing a more than 80-fold enrichment. Several predicted interactors were validated biochemically. To demonstrate generalized applicability, we used our workflow to predict protein-protein interactions dependent upon motif-specific arginine methylation. Our data demonstrate the applicability of our approach to, conceivably, any modular binding domain that recognizes a specific post-translationally modified motif. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Proteomics , Adaptor Proteins, Signal Transducing , Phosphorylation , Protein Binding , Signal Transduction , src Homology Domains
13.
PLoS One ; 13(1): e0190757, 2018.
Article in English | MEDLINE | ID: mdl-29293641

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0185317.].

14.
FEBS J ; 285(1): 72-86, 2018 01.
Article in English | MEDLINE | ID: mdl-29091353

ABSTRACT

Plexins (Plxns) are semaphorin (Sema) receptors that play important signaling roles, particularly in the developing nervous system and vasculature. Sema-Plxn signaling regulates cellular processes such as cytoskeletal dynamics, proliferation, and differentiation. However, the receptor-proximal signaling mechanisms driving Sema-Plxn signal transduction are only partially understood. Plxn tyrosine phosphorylation is thought to play an important role in these signaling events as receptor and nonreceptor tyrosine kinases have been shown to interact with Plxn receptors. The Src family kinase Fyn can induce the tyrosine phosphorylation of PlxnA1 and PlxnA2. However, the Fyn-dependent phosphorylation sites on these receptors have not been identified. Here, using mass spectrometry-based approaches, we have identified highly conserved, Fyn-induced PlexinA (PlxnA) tyrosine phosphorylation sites. Mutation of these sites to phenylalanine results in significantly decreased Fyn-dependent PlxnA tyrosine phosphorylation. Furthermore, in contrast to wild-type human PLXNA2 mRNA, mRNA harboring these point mutations cannot rescue eye developmental defects when coinjected with a plxnA2 morpholino in zebrafish embryos. Together these data suggest that Fyn-dependent phosphorylation at two critical tyrosines is a key feature of vertebrate PlxnA1 and PlxnA2 signal transduction.


Subject(s)
Eye/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Cell Surface/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Eye/embryology , HEK293 Cells , Humans , Mutation, Missense , Nerve Tissue Proteins/genetics , Phosphorylation , Receptors, Cell Surface/genetics , Sequence Homology, Amino Acid , Signal Transduction/genetics , Tyrosine/genetics , Zebrafish
15.
Gene Expr Patterns ; 27: 56-66, 2018 01.
Article in English | MEDLINE | ID: mdl-29107805

ABSTRACT

Plexins (Plxns) and Semaphorins (Semas) are key signaling molecules that regulate many aspects of development. Plxns are a family of transmembrane protein receptors that are activated upon extracellular binding by Semas. Activated Plxns trigger intracellular signaling cascades, which regulate a range of developmental processes, including axon guidance, neuronal positioning and vasculogenesis. Semas are a large family of both transmembrane and secreted signaling molecules, and show subtype specific binding to different Plxn family members. Each Plxn can play different roles in development, and so tightly regulated temporal and spatial expression of receptor subtypes is critical to ensure appropriate signaling. Here we elucidate the expression profiles of the plxnA family, plxnA1a, A1b, A2, A3 and A4 at 18, 24, 36, 48, 60 and 72 h post fertilization in the developing zebrafish. We show that PlxnA family members are expressed in neuronal tissues during zebrafish development, but exhibit key differences in expression within these tissues. We also highlight that plxnA1 has two genes in zebrafish, A1a and A1b, which show divergences in expression patterns during early development.


Subject(s)
Cell Adhesion Molecules/metabolism , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Zebrafish/genetics , Animals , Cell Adhesion Molecules/genetics , Cells, Cultured , In Situ Hybridization , Nerve Tissue Proteins/genetics , Neurons/cytology , Phylogeny , Signal Transduction , Zebrafish/metabolism , Zebrafish Proteins/genetics
16.
Biochem J ; 474(23): 3963-3984, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29025973

ABSTRACT

Discoidin, CUB, and LCCL domain containing 2 (DCBLD2) is a neuropilin-like transmembrane scaffolding receptor with known and anticipated roles in vascular remodeling and neuronal positioning. DCBLD2 is also up-regulated in several cancers and can drive glioblastomas downstream of activated epidermal growth factor receptor. While a few studies have shown either a positive or negative role for DCBLD2 in regulating growth factor receptor signaling, little is known about the conserved signaling features of DCBLD family members that drive their molecular activities. We previously identified DCBLD2 tyrosine phosphorylation sites in intracellular YxxP motifs that are required for the phosphorylation-dependent binding of the signaling adaptors CRK and CRKL (CT10 regulator of kinase and CRK-like). These intracellular YxxP motifs are highly conserved across vertebrates and between DCBLD family members. Here, we demonstrate that, as for DCBLD2, DCBLD1 YxxP motifs are required for CRKL-SH2 (Src homology 2) binding. We report that Src family kinases (SFKs) and Abl differentially promote the interaction between the CRKL-SH2 domain and DCBLD1 and DCBLD2, and while SFKs and Abl each promote DCBLD1 and DCBLD2 binding to the CRKL-SH2 domain, the effect of Abl is more pronounced for DCBLD1. Using high-performance liquid chromatography coupled with tandem mass spectrometry, we quantified phosphorylation at several YxxP sites in DCBLD1 and DCBLD2, mapping site-specific preferences for SFKs and Abl. Together, these data provide a platform to decipher the signaling mechanisms by which these novel receptors drive their biological activities.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Membrane Proteins/chemistry , Nuclear Proteins/chemistry , Oncogene Proteins v-abl/chemistry , Proto-Oncogene Proteins c-fyn/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , Conserved Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins v-abl/metabolism , Phosphorylation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Zebrafish
17.
PLoS One ; 12(9): e0185317, 2017.
Article in English | MEDLINE | ID: mdl-28934368

ABSTRACT

Histidyl tRNA Synthetase (HARS) is a member of the aminoacyl tRNA synthetase (ARS) family of enzymes. This family of 20 enzymes is responsible for attaching specific amino acids to their cognate tRNA molecules, a critical step in protein synthesis. However, recent work highlighting a growing number of associations between ARS genes and diverse human diseases raises the possibility of new and unexpected functions in this ancient enzyme family. For example, mutations in HARS have been linked to two different neurological disorders, Usher Syndrome Type IIIB and Charcot Marie Tooth peripheral neuropathy. These connections raise the possibility of previously undiscovered roles for HARS in metazoan development, with alterations in these functions leading to complex diseases. In an attempt to establish Danio rerio as a model for studying HARS functions in human disease, we characterized the Danio rerio hars gene and compared it to that of human HARS. Using a combination of bioinformatics, molecular biology, and cellular approaches, we found that while the human genome encodes separate genes for cytoplasmic and mitochondrial HARS protein, the Danio rerio genome encodes a single hars gene which undergoes alternative splicing to produce the respective cytoplasmic and mitochondrial versions of Hars. Nevertheless, while the HARS genes of humans and Danio differ significantly at the genomic level, we found that they are still highly conserved at the amino acid level, underscoring the potential utility of Danio rerio as a model organism for investigating HARS function and its link to human diseases in vivo.


Subject(s)
Cytoplasm/enzymology , Cytoplasm/genetics , Histidine-tRNA Ligase/genetics , Mitochondria/enzymology , Zebrafish/genetics , Animals , COS Cells , Chlorocebus aethiops , Conserved Sequence , Gene Expression Regulation, Enzymologic , Histidine-tRNA Ligase/chemistry , Histidine-tRNA Ligase/metabolism , Humans , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Species Specificity
18.
Dev Dyn ; 246(7): 539-549, 2017 07.
Article in English | MEDLINE | ID: mdl-28440030

ABSTRACT

BACKGROUND: Semaphorin (Sema)/Plexin (Plxn) signaling is important for many aspects of neuronal development, however, the transcriptional regulation imposed by this signaling pathway is unknown. Previously, we identified an essential role for Sema6A/PlxnA2 signaling in regulating proliferation and cohesion of retinal precursor cells (RPCs) during early eye development. This study used RNA isolated from control, Sema6A-deficient and PlxnA2-deficient zebrafish embryos in a microarray analysis to identify genes that were differentially expressed when this signaling pathway was disrupted. RESULTS: We uncovered a set of 58 transcripts, and all but 1 were up-regulated in both sema6A and plxnA2 morphants. We validated gene expression changes in subset of candidates that are suggested to be involved in proliferation, migration or neuronal positioning. We further functionally evaluated one gene, rasl11b, as contributing to disrupted proliferation in sema6A and plxna2 morphants. Our results suggest rasl11b negatively regulates proliferation of RPCs in the developing zebrafish eye. CONCLUSIONS: Microarray analysis has generated a resource of target genes downstream of Sema6A/PlxnA2 signaling, which can be further investigated to elucidate the downstream effects of this well-studied neuronal and vascular guidance signaling pathway. Developmental Dynamics 246:539-549, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Animals , Cell Movement , Cell Proliferation , Eye/embryology , Eye/growth & development , Gene Expression Regulation, Developmental/genetics , Retina/cytology , Stem Cells , Zebrafish
19.
Sci Rep ; 5: 13160, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26271225

ABSTRACT

Aminoacyl-tRNA synthetases (AARSs) catalyze an early step in protein synthesis, but also regulate diverse physiological processes in animal cells. These include angiogenesis, and human threonyl-tRNA synthetase (TARS) represents a potent pro-angiogenic AARS. Angiogenesis stimulation can be blocked by the macrolide antibiotic borrelidin (BN), which exhibits a broad spectrum toxicity that has discouraged deeper investigation. Recently, a less toxic variant (BC194) was identified that potently inhibits angiogenesis. Employing biochemical, cell biological, and biophysical approaches, we demonstrate that the toxicity of BN and its derivatives is linked to its competition with the threonine substrate at the molecular level, which stimulates amino acid starvation and apoptosis. By separating toxicity from the inhibition of angiogenesis, a direct role for TARS in vascular development in the zebrafish could be demonstrated. Bioengineered natural products are thus useful tools in unmasking the cryptic functions of conventional enzymes in the regulation of complex processes in higher metazoans.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Angiogenesis Inhibitors/administration & dosage , Angiogenic Proteins/metabolism , Macrolides/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Angiogenesis Inhibitors/chemistry , Animals , Dose-Response Relationship, Drug , Enzyme Activation , Zebrafish
20.
Development ; 141(12): 2473-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24917502

ABSTRACT

Organs are generated from collections of cells that coalesce and remain together as they undergo a series of choreographed movements to give the organ its final shape. We know little about the cellular and molecular mechanisms that regulate tissue cohesion during morphogenesis. Extensive cell movements underlie eye development, starting with the eye field separating to form bilateral vesicles that go on to evaginate from the forebrain. What keeps eye cells together as they undergo morphogenesis and extensive proliferation is unknown. Here, we show that plexina2 (Plxna2), a member of a receptor family best known for its roles in axon and cell guidance, is required alongside the repellent semaphorin 6a (Sema6a) to keep cells integrated within the zebrafish eye vesicle epithelium. sema6a is expressed throughout the eye vesicle, whereas plxna2 is restricted to the ventral vesicle. Knockdown of Plxna2 or Sema6a results in a loss of vesicle integrity, with time-lapse microscopy showing that eye progenitors either fail to enter the evaginating vesicles or delaminate from the eye epithelium. Explant experiments, and rescue of eye vesicle integrity with simultaneous knockdown of sema6a and plxna2, point to an eye-autonomous requirement for Sema6a/Plxna2. We propose a novel, tissue-autonomous mechanism of organ cohesion, with neutralization of repulsion suggested as a means to promote interactions between cells within a tissue domain.


Subject(s)
Eye/embryology , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/physiology , Receptors, Cell Surface/physiology , Semaphorins/physiology , Zebrafish Proteins/physiology , Animals , Axons/metabolism , Cell Communication , Cell Movement , Cell Proliferation , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Morphogenesis , Nerve Tissue Proteins/genetics , Prosencephalon/embryology , Receptors, Cell Surface/genetics , Semaphorins/genetics , Signal Transduction , Stem Cells/cytology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...