Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nature ; 591(7849): 281-287, 2021 03.
Article in English | MEDLINE | ID: mdl-33568815

ABSTRACT

Skeletal muscle regenerates through the activation of resident stem cells. Termed satellite cells, these normally quiescent cells are induced to proliferate by wound-derived signals1. Identifying the source and nature of these cues has been hampered by an inability to visualize the complex cell interactions that occur within the wound. Here we use muscle injury models in zebrafish to systematically capture the interactions between satellite cells and the innate immune system after injury, in real time, throughout the repair process. This analysis revealed that a specific subset of macrophages 'dwell' within the injury, establishing a transient but obligate niche for stem cell proliferation. Single-cell profiling identified proliferative signals that are secreted by dwelling macrophages, which include the cytokine nicotinamide phosphoribosyltransferase (Nampt, which is also known as visfatin or PBEF in humans). Nampt secretion from the macrophage niche is required for muscle regeneration, acting through the C-C motif chemokine receptor type 5 (Ccr5), which is expressed on muscle stem cells. This analysis shows that in addition to their ability to modulate the immune response, specific macrophage populations also provide a transient stem-cell-activating niche, directly supplying proliferation-inducing cues that govern the repair process that is mediated by muscle stem cells. This study demonstrates that macrophage-derived niche signals for muscle stem cells, such as NAMPT, can be applied as new therapeutic modalities for skeletal muscle injury and disease.


Subject(s)
Macrophages/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Myoblasts/cytology , Nicotinamide Phosphoribosyltransferase/metabolism , Stem Cell Niche , Zebrafish/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Humans , Macrophages/cytology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , PAX7 Transcription Factor/metabolism , RNA-Seq , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Regeneration/physiology , Single-Cell Analysis , Zebrafish/immunology
2.
PLoS Genet ; 18(6): e1010287, 2022 06.
Article in English | MEDLINE | ID: mdl-35737712

ABSTRACT

Myofibrils of the skeletal muscle are comprised of sarcomeres that generate force by contraction when myosin-rich thick filaments slide past actin-based thin filaments. Surprisingly little is known about the molecular processes that guide sarcomere assembly in vivo, despite deficits within this process being a major cause of human disease. To overcome this knowledge gap, we undertook a forward genetic screen coupled with reverse genetics to identify genes required for vertebrate sarcomere assembly. In this screen, we identified a zebrafish mutant with a nonsense mutation in mob4. In Drosophila, mob4 has been reported to play a role in spindle focusing as well as neurite branching and in planarians mob4 was implemented in body size regulation. In contrast, zebrafish mob4geh mutants are characterised by an impaired actin biogenesis resulting in sarcomere defects. Whereas loss of mob4 leads to a reduction in the amount of myofibril, transgenic expression of mob4 triggers an increase. Further genetic analysis revealed the interaction of Mob4 with the actin-folding chaperonin TRiC, suggesting that Mob4 impacts on TRiC to control actin biogenesis and thus myofibril growth. Additionally, mob4geh features a defective microtubule network, which is in-line with tubulin being the second main folding substrate of TRiC. We also detected similar characteristics for strn3-deficient mutants, which confirmed Mob4 as a core component of STRIPAK and surprisingly implicates a role of the STRIPAK complex in sarcomerogenesis.


Subject(s)
Myofibrils , Zebrafish , Actins/genetics , Actins/metabolism , Animals , Chaperonins/metabolism , Microtubules/genetics , Myofibrils/metabolism , Sarcomeres/metabolism , Zebrafish/genetics
3.
PLoS Genet ; 18(2): e1010066, 2022 02.
Article in English | MEDLINE | ID: mdl-35148320

ABSTRACT

Myofibrils within skeletal muscle are composed of sarcomeres that generate force by contraction when their myosin-rich thick filaments slide past actin-based thin filaments. Although mutations in components of the sarcomere are a major cause of human disease, the highly complex process of sarcomere assembly is not fully understood. Current models of thin filament assembly highlight a central role for filament capping proteins, which can be divided into three protein families, each ascribed with separate roles in thin filament assembly. CapZ proteins have been shown to bind the Z-disc protein α-actinin to form an anchoring complex for thin filaments and actin polymerisation. Subsequent thin filaments extension dynamics are thought to be facilitated by Leiomodins (Lmods) and thin filament assembly is concluded by Tropomodulins (Tmods) that specifically cap the pointed end of thin filaments. To study thin filament assembly in vivo, single and compound loss-of-function zebrafish mutants within distinct classes of capping proteins were analysed. The generated lmod3- and capza1b-deficient zebrafish exhibited aspects of the pathology caused by variations in their human orthologs. Although loss of the analysed main capping proteins of the skeletal muscle, capza1b, capza1a, lmod3 and tmod4, resulted in sarcomere defects, residual organised sarcomeres were formed within the assessed mutants, indicating that these proteins are not essential for the initial myofibril assembly. Furthermore, detected similarity and location of myofibril defects, apparent at the peripheral ends of myofibres of both Lmod3- and CapZα-deficient mutants, suggest a function in longitudinal myofibril growth for both proteins, which is molecularly distinct to the function of Tmod4.


Subject(s)
CapZ Actin Capping Protein/metabolism , Muscular Diseases , Myofibrils , Actins/genetics , Actins/metabolism , Animals , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Diseases/genetics , Muscular Diseases/metabolism , Myofibrils/genetics , Myofibrils/metabolism , Tropomodulin/genetics , Tropomodulin/metabolism , Zebrafish/genetics , Zebrafish/metabolism
4.
Nature ; 535(7613): 542-6, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27437584

ABSTRACT

The transition from fins to limbs was an important terrestrial adaptation, but how this crucial evolutionary shift arose developmentally is unknown. Current models focus on the distinct roles of the apical ectodermal ridge (AER) and the signaling molecules that it secretes during limb and fin outgrowth. In contrast to the limb AER, the AER of the fin rapidly transitions into the apical fold and in the process shuts off AER-derived signals that stimulate proliferation of the precursors of the appendicular skeleton. The differing fates of the AER during fish and tetrapod development have led to the speculation that fin-fold formation was one of the evolutionary hurdles to the AER-dependent expansion of the fin mesenchyme required to generate the increased appendicular structure evident within limbs. Consequently, a heterochronic shift in the AER-to-apical-fold transition has been postulated to be crucial for limb evolution. The ability to test this model has been hampered by a lack of understanding of the mechanisms controlling apical fold induction. Here we show that invasion by cells of a newly identified somite-derived lineage into the AER in zebrafish regulates apical fold induction. Ablation of these cells inhibits apical fold formation, prolongs AER activity and increases the amount of fin bud mesenchyme, suggesting that these cells could provide the timing mechanism proposed in Thorogood's clock model of the fin-to-limb transition. We further demonstrate that apical-fold inducing cells are progressively lost during gnathostome evolution;the absence of such cells within the tetrapod limb suggests that their loss may have been a necessary prelude to the attainment of limb-like structures in Devonian sarcopterygian fish.


Subject(s)
Animal Fins/embryology , Animal Fins/metabolism , Ectoderm/embryology , Ectoderm/metabolism , Somites/embryology , Somites/metabolism , Zebrafish/embryology , Animals , Biological Evolution , Cell Lineage , Ectoderm/cytology , Female , Limb Buds/cytology , Limb Buds/embryology , Limb Buds/metabolism , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Somites/cytology
5.
J Cell Mol Med ; 24(12): 6680-6689, 2020 06.
Article in English | MEDLINE | ID: mdl-32343037

ABSTRACT

Duchenne muscular dystrophy is a severe muscle wasting disease caused by mutations in the dystrophin gene (dmd). Ataluren has been approved by the European Medicines Agency for treatment of Duchenne muscular dystrophy. Ataluren has been reported to promote ribosomal read-through of premature stop codons, leading to restoration of full-length dystrophin protein. However, the mechanism of Ataluren action has not been fully described. To evaluate the efficacy of Ataluren on all three premature stop codons featuring different termination strengths (UAA > UAG > UGA), novel dystrophin-deficient zebrafish were generated. Pathological assessment of the muscle by birefringence quantification, a tool to directly measure muscle integrity, did not reveal a significant effect of Ataluren on any of the analysed dystrophin-deficient mutants at 3 days after fertilization. Functional analysis of the musculature at 6 days after fertilization by direct measurement of the generated force revealed a significant improvement by Ataluren only for the UAA-carrying mutant dmdta222a . Interestingly however, all other analysed dystrophin-deficient mutants were not affected by Ataluren, including the dmdpc3 and dmdpc2 mutants that harbour weaker premature stop codons UAG and UGA, respectively. These in vivo results contradict reported in vitro data on Ataluren efficacy, suggesting that Ataluren might not promote read-through of premature stop codons. In addition, Ataluren had no effect on dystrophin transcript levels, but mild adverse effects on wild-type larvae were identified. Further assessment of N-terminally truncated dystrophin opened the possibility of Ataluren promoting alternative translation codons within dystrophin, thereby potentially shifting the patient cohort applicable for Ataluren.


Subject(s)
Dystrophin/genetics , Mutation/genetics , Oxadiazoles/pharmacology , Animals , Codon, Nonsense/genetics , Exons/genetics , Homozygote , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Oxadiazoles/adverse effects , Phenotype , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/genetics
6.
Nature ; 512(7514): 314-8, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25119043

ABSTRACT

Haematopoietic stem cells (HSCs) are self-renewing stem cells capable of replenishing all blood lineages. In all vertebrate embryos that have been studied, definitive HSCs are generated initially within the dorsal aorta (DA) of the embryonic vasculature by a series of poorly understood inductive events. Previous studies have identified that signalling relayed from adjacent somites coordinates HSC induction, but the nature of this signal has remained elusive. Here we reveal that somite specification of HSCs occurs via the deployment of a specific endothelial precursor population, which arises within a sub-compartment of the zebrafish somite that we have defined as the endotome. Endothelial cells of the endotome are specified within the nascent somite by the activity of the homeobox gene meox1. Specified endotomal cells consequently migrate and colonize the DA, where they induce HSC formation through the deployment of chemokine signalling activated in these cells during endotome formation. Loss of meox1 activity expands the endotome at the expense of a second somitic cell type, the muscle precursors of the dermomyotomal equivalent in zebrafish, the external cell layer. The resulting increase in endotome-derived cells that migrate to colonize the DA generates a dramatic increase in chemokine-dependent HSC induction. This study reveals the molecular basis for a novel somite lineage restriction mechanism and defines a new paradigm in induction of definitive HSCs.


Subject(s)
Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Somites/cytology , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Animals , Aorta/cytology , Aorta/embryology , Biomarkers/analysis , Cell Movement , Chemokine CXCL12/analysis , Chemokine CXCL12/metabolism , Chick Embryo , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/analysis , Homeodomain Proteins/genetics , Humans , Mice , Muscles/cytology , Muscles/metabolism , Mutation/genetics , Somites/metabolism , Transcription Factors/analysis , Transcription Factors/genetics , Wnt Proteins/analysis , Wnt Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/analysis , Zebrafish Proteins/genetics
7.
Hum Mol Genet ; 26(6): 1146-1156, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28104788

ABSTRACT

Congenital myopathies are muscle degenerative disorders with a broad clinical spectrum. A number of myopathies have been associated with molecular defects within sarcomeres, the force-generating component of the muscle cell. Whereas the highly regular organization of the myofibril has been studied in detail, in vivo assembly of sarcomeres remains a poorly understood process. Therefore, a more detailed knowledge of sarcomere assembly is crucial to better understand the pathogenic mechanisms within myopathies. Recently, mutations in myosin XVIIIB (MYO18B) have been associated with cases of myopathies, although the underlying mechanism for the resulting pathology remains to be defined. To analyze the role of myosin XVIIIB in skeletal muscle disease, zebrafish mutants for myo18b were generated. Full loss of myo18b function results in a complete lack of sarcomeric structure, revealing a highly surprising and essential role for myo18b in sarcomere assembly. Importantly, scattered thin and thick filaments assemble throughout the sarcoplasm; but fail to organize into recognizable sarcomeric structures in myo18b null mutants. In myo18b partial loss-of-function mutants sarcomeric structures are assembled, but thin and thick filaments remain misaligned within these structures. These observations suggest a novel model of sarcomere assembly where Myo18b coordinates the integration of preformed thick and thin filaments into the sarcomere. Disruption of this highly coordinated process results in a block in sarcomere biogenesis and the onset of myopathic pathology.


Subject(s)
Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Myosins/genetics , Sarcomeres/genetics , Tumor Suppressor Proteins/genetics , Zebrafish/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Humans , Muscle, Skeletal/pathology , Mutant Proteins/genetics , Myopathies, Structural, Congenital/pathology , Myosins/biosynthesis , Sarcomeres/metabolism , Sarcomeres/pathology , Tumor Suppressor Proteins/biosynthesis , Zebrafish/physiology
8.
Dev Dyn ; 246(12): 1027-1035, 2017 12.
Article in English | MEDLINE | ID: mdl-28791777

ABSTRACT

BACKGROUND: The microtubule-severing protein complex katanin is composed two subunits, the ATPase subunit, KATNA1, and the noncatalytic regulatory subunit, KATNB1. Recently, the Katnb1 gene has been linked to infertility, regulation of centriole and cilia formation in fish and mammals, as well as neocortical brain development. KATNB1 protein is expressed in germ cells in humans and mouse, mitotic/meiotic spindles and cilia, although the full expression pattern of the Katnb1 gene has not been described. RESULTS: Using a knockin-knockout mouse model of Katnb1 dysfunction we demonstrate that Katnb1 is ubiquitously expressed during embryonic development, although a stronger expression is seen in the crown cells of the gastrulation organizer, the murine node. Furthermore, null and hypomorphic Katnb1 gene mutations show a novel correlation between Katnb1 dysregulation and the development of impaired left-right signaling, including cardiac malformations. CONCLUSIONS: Katanin function is a critical regulator of heart development in mice. These findings are potentially relevant to human cardiac development. Developmental Dynamics 246:1027-1035, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Heart Defects, Congenital , Katanin , Mutation , Signal Transduction/genetics , Animals , Gene Knock-In Techniques , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Katanin/genetics , Katanin/metabolism , Mice , Mice, Knockout
9.
Differentiation ; 91(1-3): 29-41, 2016.
Article in English | MEDLINE | ID: mdl-26897459

ABSTRACT

Nkx2-5 is one of the master regulators of cardiac development, homeostasis and disease. This transcription factor has been previously associated with a suite of cardiac congenital malformations and impairment of electrical activity. When disease causative mutations in transcription factors are considered, NKX2-5 gene dysfunction is the most common abnormality found in patients. Here we describe a novel mouse model and subsequent implications of Nkx2-5 loss for aspects of myocardial electrical activity. In this work we have engineered a new Nkx2-5 conditional knockout mouse in which flox sites flank the entire Nkx2-5 locus, and validated this line for the study of heart development, differentiation and disease using a full deletion strategy. While our homozygous knockout mice show typical embryonic malformations previously described for the lack of the Nkx2-5 gene, hearts of heterozygous adult mice show moderate morphological and functional abnormalities that are sufficient to sustain blood supply demands under homeostatic conditions. This study further reveals intriguing aspects of Nkx2-5 function in the control of cardiac electrical activity. Using a combination of mouse genetics, biochemistry, molecular and cell biology, we demonstrate that Nkx2-5 regulates the gene encoding Kcnh2 channel and others, shedding light on potential mechanisms generating electrical abnormalities observed in patients bearing NKX2-5 dysfunction and opening opportunities to the study of novel therapeutic targets for anti-arrhythmogenic therapies.


Subject(s)
ERG1 Potassium Channel/genetics , Heart Defects, Congenital/genetics , Heart/growth & development , Homeobox Protein Nkx-2.5/genetics , Animals , Disease Models, Animal , ERG1 Potassium Channel/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Heart/physiopathology , Heart Defects, Congenital/physiopathology , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, Knockout , Mutation
10.
PLoS Biol ; 9(10): e1001168, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21990962

ABSTRACT

Locomotor strategies in terrestrial tetrapods have evolved from the utilisation of sinusoidal contractions of axial musculature, evident in ancestral fish species, to the reliance on powerful and complex limb muscles to provide propulsive force. Within tetrapods, a hindlimb-dominant locomotor strategy predominates, and its evolution is considered critical for the evident success of the tetrapod transition onto land. Here, we determine the developmental mechanisms of pelvic fin muscle formation in living fish species at critical points within the vertebrate phylogeny and reveal a stepwise modification from a primitive to a more derived mode of pelvic fin muscle formation. A distinct process generates pelvic fin muscle in bony fishes that incorporates both primitive and derived characteristics of vertebrate appendicular muscle formation. We propose that the adoption of the fully derived mode of hindlimb muscle formation from this bimodal character state is an evolutionary innovation that was critical to the success of the tetrapod transition.


Subject(s)
Animal Fins/growth & development , Biological Evolution , Fishes/growth & development , Muscle Development , Pelvis/growth & development , Animal Fins/anatomy & histology , Animals , Animals, Genetically Modified , Fishes/genetics , Pelvis/anatomy & histology , Phylogeny , Somites/transplantation , Species Specificity
11.
Dev Biol ; 368(2): 193-202, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22609552

ABSTRACT

The Hedgehog (HH) signaling pathway is a central regulator of embryonic development, controlling the pattern and proliferation of a wide variety of organs. Previous studies have implicated the secreted protein, Scube2, in HH signal transduction in the zebrafish embryo (Hollway et al., 2006; Kawakami et al., 2005; Woods and Talbot, 2005) although the nature of the molecular function of Scube2 in this process has remained undefined. This analysis has been compounded by the fact that removal of Scube2 activity in the zebrafish embryo leads to only subtle defects in HH signal transduction in vivo (Barresi et al., 2000; Hollway et al., 2006; Ochi and Westerfield, 2007; van Eeden et al., 1996; Wolff et al., 2003). Here we present the discovery of two additional scube genes in zebrafish, scube1 and scube3, and demonstrate their roles in facilitating HH signal transduction. Knocking down the function of all three scube genes simultaneously phenocopies a complete loss of HH signal transduction in the embryo, revealing that Scube signaling is essential for HH signal transduction in vivo. We further define the molecular role of scube2 in HH signaling.


Subject(s)
Calcium-Binding Proteins/genetics , Embryo, Nonmammalian/metabolism , Extracellular Matrix Proteins/genetics , Hedgehog Proteins/genetics , Signal Transduction/genetics , Zebrafish Proteins/genetics , Animals , Blotting, Western , COS Cells , Calcium-Binding Proteins/metabolism , Chlorocebus aethiops , DNA, Complementary/chemistry , DNA, Complementary/genetics , Embryo, Nonmammalian/embryology , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , In Situ Hybridization , Molecular Sequence Data , Multigene Family , Mutation , Phenotype , Sequence Analysis, DNA , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism
12.
J Ophthalmic Vis Res ; 18(1): 60-67, 2023.
Article in English | MEDLINE | ID: mdl-36937198

ABSTRACT

Purpose: Mutations in TCP-1 ring complex (TRiC) have been associated with Leber Congenital Amaurosis (LCA). TRiC is involved in protein folding and has 8 essential subunits including CCT5. Herein, we studied the retina of TRiC mutant zebrafish to evaluate the possible role of impaired actin and tubulin folding in LCA. Methods: The cct5 t f 212 b retina was histologically studied using Toluidine Blue staining as well as TUNEL, BrdU-labeling, and Phalloidin assays. Retinal organisation was assessed by quantification of the cellularity utilising DAPI. Results: Laminar organization of cct5 t f 212 b retinas was intact. Enhanced apoptosis throughout the cct5 t f 212 b retina was not compensated by higher proliferation rates, leaving the cct5 t f 212 b retina smaller in size. Quantification of retinal layer cellularity demonstrated that specifically the numbers of the amacrine and the retinal ganglion cells were depleted, suggesting that the cct5 t f 212 b retina was not uniformly affected by the reduced actin folding. Conclusion: Whereas the current literature suggests that LCA is predominantly affecting retinal photoreceptor cells and the retinal pigment epithelium, cct5 t f 212 b analyses demonstrated the important role of folding of actin by TRiC, suggesting that cct5 t f 212 b is a useful tool to specifically analyze the role of F-actin filaments in the context of LCA.

13.
Dev Cell ; 12(2): 207-19, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17276339

ABSTRACT

Somites are transient, mesodermally derived structures that give rise to a number of different cell types within the vertebrate embryo. To achieve this, somitic cells are partitioned into lineage-restricted domains, whose fates are determined by signals secreted from adjacent tissues. While the molecular nature of many of the inductive signals that trigger formation of different cell fates within the nascent somite has been identified, less is known about the processes that coordinate the formation of the subsomitic compartments from which these cells arise. Utilizing a combination of vital dye-staining and lineage-tracking techniques, we describe a previously uncharacterized, lineage-restricted compartment of the zebrafish somite that generates muscle progenitor cells for the growth of appendicular, hypaxial, and axial muscles during development. We also show that formation of this compartment occurs via whole-somite rotation, a process that requires the action of the Sdf family of secreted cytokines.


Subject(s)
Body Patterning/physiology , Cell Compartmentation , Embryo, Nonmammalian/cytology , Muscle Cells/cytology , Somites/physiology , Stem Cells/cytology , Zebrafish/embryology , Animals , Cell Lineage , Embryo, Nonmammalian/embryology , Gene Expression Regulation, Developmental , Muscle Cells/metabolism , PAX7 Transcription Factor/metabolism , Rotation , Signal Transduction , Somites/cytology , Stem Cells/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Development ; 136(19): 3367-76, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19736328

ABSTRACT

The skeletal muscle basement membrane fulfils several crucial functions during development and in the mature myotome and defects in its composition underlie certain forms of muscular dystrophy. A major component of this extracellular structure is the laminin polymer, which assembles into a resilient meshwork that protects the sarcolemma during contraction. Here we describe a zebrafish mutant, softy, which displays severe embryonic muscle degeneration as a result of initial basement membrane failure. The softy phenotype is caused by a mutation in the lamb2 gene, identifying laminin beta2 as an essential component of this basement membrane. Uniquely, softy homozygotes are able to recover and survive to adulthood despite the loss of myofibre adhesion. We identify the formation of ectopic, stable basement membrane attachments as a novel means by which detached fibres are able to maintain viability. This demonstration of a muscular dystrophy model possessing innate fibre viability following muscle detachment suggests basement membrane augmentation as a therapeutic strategy to inhibit myofibre loss.


Subject(s)
Laminin/genetics , Laminin/physiology , Muscular Dystrophy, Animal/embryology , Muscular Dystrophy, Animal/genetics , Mutation , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology , Zebrafish/embryology , Zebrafish/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Basement Membrane/pathology , Cell Survival , DNA Primers/genetics , Eye/embryology , Homozygote , Molecular Sequence Data , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Sarcolemma/pathology , Sequence Homology, Amino Acid
15.
Dev Dyn ; 240(2): 422-31, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21246659

ABSTRACT

Laminins are essential components of all basement membranes and are fundamental to tissue development and homeostasis. Humans possess at least 16 different heterotrimeric laminin complexes formed through different combinations of alpha, beta, and gamma chains. Individual chains appear to exhibit unique expression patterns, leading to the notion that overlap between expression domains governs the constitution of complexes found within particular tissues. However, the spatial and temporal expression of laminin genes has not been comprehensively analyzed in any vertebrate model to date. Here, we describe the tissue-specific expression patterns of all laminin genes in the zebrafish, throughout embryonic development and into the "post-juvenile" period, which is representative of the adult body form. In addition, we present phylogenetic and microsynteny analyses, which demonstrate that the majority of our zebrafish sequences are orthologous to human laminin genes. Together, these data represent a fundamental resource for the study of vertebrate laminins.


Subject(s)
Biological Evolution , Gene Expression Regulation, Developmental , Laminin/genetics , Multigene Family , Protein Isoforms/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Humans , In Situ Hybridization , Laminin/classification , Laminin/metabolism , Phylogeny , Protein Isoforms/classification , Protein Isoforms/metabolism , Synteny , Tissue Distribution , Zebrafish/anatomy & histology
16.
J Cell Mol Med ; 15(12): 2643-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21251213

ABSTRACT

Duchenne muscular dystophy (DMD) is a severe muscle wasting disease caused by mutations in the dystrophin gene. By utilizing antisense oligonucleotides, splicing of the dystrophin transcript can be altered so that exons harbouring a mutation are excluded from the mature mRNA. Although this approach has been shown to be effective to restore partially functional dystrophin protein, the level of dystrophin protein that is necessary to rescue a severe muscle pathology has not been addressed. As zebrafish dystrophin mutants (dmd) resemble the severe muscle pathology of human patients, we have utilized this model to evaluate exon skipping. Novel dmd mutations were identified to enable the design of phenotype rescue studies via morpholino administration. Correlation of induced exon-skipping efficiency and the level of phenotype rescue suggest that relatively robust levels of exon skipping are required to achieve significant therapeutic ameliorations and that pre-screening analysis of exon-skipping drugs in zebrafish may help to more accurately predict clinical trials for therapies of DMD.


Subject(s)
Dystrophin/physiology , Exons/genetics , Muscular Dystrophy, Duchenne/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Humans , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Zebrafish/growth & development , Zebrafish/metabolism
17.
BMC Biol ; 5: 34, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17705855

ABSTRACT

BACKGROUND: Over the last two decades, zebrafish have been established as a genetically versatile model system for investigating many different aspects of vertebrate developmental biology. With the credentials of zebrafish as a developmental model now well recognized, the emerging new opportunity is the wider application of zebrafish biology to aspects of human disease modelling. This rapidly increasing use of zebrafish as a model for human disease has necessarily generated interest in the anatomy of later developmental phases such as the larval, juvenile, and adult stages, during which many of the key aspects of organ morphogenesis and maturation take place. Anatomical resources and references that encompass these stages are non-existent in zebrafish and there is therefore an urgent need to understand how different organ systems and anatomical structures develop throughout the life of the fish. RESULTS: To overcome this deficit we have utilized the technique of optical projection tomography to produce three-dimensional (3D) models of larval fish. In order to view and display these models we have created FishNet http://www.fishnet.org.au, an interactive reference of zebrafish anatomy spanning the range of zebrafish development from 24 h until adulthood. CONCLUSION: FishNet contains more than 36,000 images of larval zebrafish, with more than 1,500 of these being annotated. The 3D models can be manipulated on screen or virtually sectioned. This resource represents the first complete embryo to adult atlas for any species in 3D.


Subject(s)
Databases, Factual , Online Systems , Zebrafish/anatomy & histology , Animals , Embryo, Nonmammalian/anatomy & histology , Imaging, Three-Dimensional , Larva/anatomy & histology , Zebrafish/embryology
18.
Cell Rep ; 22(2): 313-322, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29320728

ABSTRACT

The TCP-1 ring complex (TRiC) is a multi-subunit group II chaperonin that assists nascent or misfolded proteins to attain their native conformation in an ATP-dependent manner. Functional studies in yeast have suggested that TRiC is an essential and generalized component of the protein-folding machinery of eukaryotic cells. However, TRiC's involvement in specific cellular processes within multicellular organisms is largely unknown because little validation of TRiC function exists in animals. Our in vivo analysis reveals a surprisingly specific role of TRiC in the biogenesis of skeletal muscle α-actin during sarcomere assembly in myofibers. TRiC acts at the sarcomere's Z-disk, where it is required for efficient assembly of actin thin filaments. Binding of ATP specifically by the TRiC subunit Cct5 is required for efficient actin folding in vivo. Furthermore, mutant α-actin isoforms that result in nemaline myopathy in patients obtain their pathogenic conformation via this function of TRiC.


Subject(s)
Actins/metabolism , Chaperonin Containing TCP-1/metabolism , Chaperonins/chemistry , Sarcomeres/metabolism , Animals , Humans , Zebrafish
19.
Proc Biol Sci ; 274(1609): 577-82, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-17476779

ABSTRACT

Tame behaviour, i.e. low wariness, in terrestrial island species is often attributed to low predation pressure. However, we know little about its physiological control and its flexibility in the face of predator introductions. Marine iguanas (Amblyrhynchus cristatus) on the Galapagos Islands are a good model to study the physiological correlates of low wariness. They have lived virtually without predation for 5-15 Myr until some populations were first confronted with feral cats and dogs some 150 years ago. We tested whether and to what extent marine iguanas can adjust their behaviour and endocrine stress response to novel predation threats. Here, we show that a corticosterone stress response to experimental chasing is absent in naive animals, but is quickly restored with experience. Initially, low wariness also increases with experience, but remains an order of magnitude too low to allow successful escape from introduced predators. Our data suggest that the ability of marine iguanas to cope with predator introductions is limited by narrow reaction norms for behavioural wariness rather than by constraints in the underlying physiological stress system. In general, we predict that island endemics show flexible physiological stress responses but are restricted by narrow behavioural plasticity.


Subject(s)
Adaptation, Physiological , Escape Reaction/physiology , Iguanas/physiology , Predatory Behavior , Stress, Physiological/physiopathology , Animals , Corticosterone/blood , Food Chain , Geography , Iguanas/blood , Male , Population Dynamics , Stress, Physiological/blood
20.
Horm Behav ; 52(5): 653-63, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17904141

ABSTRACT

For the past 5 to 15 million years, marine iguanas (Amblyrhynchus cristatus), endemic to the Galápagos archipelago, experienced relaxed predation pressure and consequently show negligible anti-predator behavior. However, over the past few decades introduced feral cats and dogs started to prey on iguanas on some of the islands. We investigated experimentally whether behavioral and endocrine anti-predator responses changed in response to predator introduction. We hypothesized that flight initiation distances (FID) and corticosterone (CORT) concentrations should increase in affected populations to cope with the novel predators. Populations of marine iguanas reacted differentially to simulated predator approach depending on whether or not they were previously naturally exposed to introduced predators. FIDs were larger at sites with predation than at sites without predation. Furthermore, the occurrence of new predators was associated with increased stress-induced CORT levels in marine iguanas. In addition, age was a strong predictor of variation in FID and CORT levels. Juveniles, which are generally more threatened by predators compared to adults, showed larger FIDs and higher CORT baseline levels as well as higher stress-induced levels than adults. The results demonstrate that this naive island species shows behavioral and physiological plasticity associated with actual predation pressure, a trait that is presumably adaptive. However, the adjustments in FID are not sufficient to cope with the novel predators. We suggest that low behavioral plasticity in the face of introduced predators may drive many island species to extinction.


Subject(s)
Behavior, Animal/physiology , Escape Reaction/physiology , Iguanas/physiology , Predatory Behavior/physiology , Social Dominance , Age Factors , Animals , Cats , Dogs , Ecuador , Female , Food Chain , Hydrocortisone/analysis , Male , Seawater
SELECTION OF CITATIONS
SEARCH DETAIL