Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Cell Rep ; 22(11): 3021-3031, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29539428

ABSTRACT

Developmental genes can have complex cis-regulatory regions with multiple enhancers. Early work revealed remarkable modularity of enhancers, whereby distinct DNA regions drive gene expression in defined spatiotemporal domains. Nevertheless, a few reports have shown that enhancers function in multiple developmental stages, implying that enhancers can be pleiotropic. Here, we have studied the activity of the enhancers of the shavenbaby gene throughout D. melanogaster development. We found that all seven shavenbaby enhancers drive expression in multiple tissues and developmental stages. We explored how enhancer pleiotropy is encoded in two of these enhancers. In one enhancer, the same transcription factor binding sites contribute to embryonic and pupal expression, revealing site pleiotropy, whereas for a second enhancer, these roles are encoded by distinct sites. Enhancer pleiotropy may be a common feature of cis-regulatory regions of developmental genes, and site pleiotropy may constrain enhancer evolution in some cases.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genetic Pleiotropy/genetics , Transcription Factors/metabolism , Humans
2.
PLoS One ; 11(1): e0146155, 2016.
Article in English | MEDLINE | ID: mdl-26727370

ABSTRACT

Quaking (QKI) is an RNA-binding protein involved in post-transcriptional mRNA processing. This gene is found to be associated with several human neurological disorders. Early expression of QKI proteins in the developing mouse neuroepithelium, together with neural tube defects in Qk mouse mutants, suggest the functional requirement of Qk for the establishment of the nervous system. As a knockout of Qk is embryonic lethal in mice, other model systems like the zebrafish could serve as a tool to study the developmental functions of qki. In the present study we sought to characterize the evolutionary relationship and spatiotemporal expression of qkia, qki2, and qkib; zebrafish homologs of human QKI. We found that qkia is an ancestral paralog of the single tetrapod Qk gene that was likely lost during the fin-to-limb transition. Conversely, qkib and qki2 are orthologs, emerging at the root of the vertebrate and teleost lineage, respectively. Both qki2 and qkib, but not qkia, were expressed in the progenitor domains of the central nervous system, similar to expression of the single gene in mice. Despite having partially overlapping expression domains, each gene has a unique expression pattern, suggesting that these genes have undergone subfunctionalization following duplication. Therefore, we suggest the zebrafish could be used to study the separate functions of qki genes during embryonic development.


Subject(s)
RNA-Binding Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Biological Evolution , Chordata/anatomy & histology , Chordata/genetics , Conserved Sequence , Evolution, Molecular , Extremities/embryology , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization, Fluorescence , Mice , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nervous System/embryology , Nervous System/metabolism , Neural Tube/metabolism , Phylogeny , RNA-Binding Proteins/biosynthesis , Real-Time Polymerase Chain Reaction , Sequence Alignment , Sequence Homology , Species Specificity , Synteny , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/biosynthesis
3.
PLoS One ; 7(9): e45175, 2012.
Article in English | MEDLINE | ID: mdl-23028827

ABSTRACT

DYT1 dystonia is caused by mutation of the TOR1A gene, resulting in the loss of a single glutamic acid residue near the carboxyl terminal of TorsinA. The neuronal functions perturbed by TorsinA[ΔE] are a major unresolved issue in understanding the pathophysiology of dystonia, presenting a critical roadblock to developing effective treatments. We identified and characterized the zebrafish homologue of TOR1A, as a first step towards elucidating the functions of TorsinA in neurons, in vivo, using the genetically-manipulable zebrafish model. The zebrafish genome was found to contain a single alternatively-spliced tor1 gene, derived from a common ancestral locus shared with the dual TOR1A and TOR1B paralogues found in tertrapods. tor1 was expressed ubiquitously during early embryonic development and in multiple adult tissues, including the CNS. The 2.1 kb tor1 mRNA encodes Torsin1, which is 59% identical and 78% homologous to human TorsinA. Torsin1 was expressed as major 45 kDa and minor 47 kDa glycoproteins, within the cytoplasm of neurons and neuropil throughout the CNS. Similar to previous findings relating to human TorsinA, mutations of the ATP hydrolysis domain of Torsin1 resulted in relocalization of the protein in cultured cells from the endoplasmic reticulum to the nuclear envelope. Zebrafish embryos lacking tor1 during early development did not show impaired viability, overt morphological abnormalities, alterations in motor behavior, or developmental defects in the dopaminergic system. Torsin1 is thus non-essential for early development of the motor system, suggesting that important CNS functions may occur later in development, consistent with the critical time window in late childhood when dystonia symptoms usually emerge in DYT1 patients. The similarities between Torsin1 and human TorsinA in domain organization, expression pattern, and cellular localization suggest that the zebrafish will provide a useful model to understand the neuronal functions of Torsins in vivo.


Subject(s)
Central Nervous System/metabolism , Gene Expression Regulation, Developmental , Molecular Chaperones/genetics , Neurons/metabolism , Zebrafish/genetics , Amino Acid Sequence , Animals , Central Nervous System/cytology , Central Nervous System/embryology , Dystonia/embryology , Dystonia/genetics , Dystonia/metabolism , Embryo, Nonmammalian , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Molecular Chaperones/metabolism , Molecular Sequence Data , Motor Activity , Mutation , Neurons/cytology , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Phylogeny , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Sequence Homology, Amino Acid , Zebrafish/metabolism
4.
J Biol Chem ; 287(5): 2971-83, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22128150

ABSTRACT

α-Synuclein is strongly implicated in the pathogenesis of Parkinson disease. However, the normal functions of synucleins and how these relate to disease pathogenesis are uncertain. We characterized endogenous zebrafish synucleins in order to develop tractable models to elucidate the physiological roles of synucleins in neurons in vivo. Three zebrafish genes, sncb, sncg1, and sncg2 (encoding ß-, γ1-, and γ2-synucleins respectively), show extensive phylogenetic conservation with respect to their human paralogues. A zebrafish α-synuclein orthologue was not found. Abundant 1.45-kb sncb and 2.7-kb sncg1 mRNAs were detected in the CNS from early development through adulthood and showed overlapping but distinct expression patterns. Both transcripts were detected in catecholaminergic neurons throughout the CNS. Zebrafish lacking ß-, γ1-, or both synucleins during early development showed normal CNS and body morphology but exhibited decreased spontaneous motor activity that resolved as gene expression recovered. Zebrafish lacking both ß- and γ1-synucleins were more severely hypokinetic than animals lacking one or the other synuclein and showed delayed differentiation of dopaminergic neurons and reduced dopamine levels. Phenotypic abnormalities resulting from loss of endogenous zebrafish synucleins were rescued by expression of human α-synuclein. These data demonstrate that synucleins have essential phylogenetically conserved neuronal functions that regulate dopamine homeostasis and spontaneous motor behavior. Zebrafish models will allow further elucidation of the molecular physiology and pathophysiology of synucleins in vivo.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , Motor Activity/physiology , Zebrafish Proteins/metabolism , beta-Synuclein/metabolism , gamma-Synuclein/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Dopamine/genetics , Humans , Immobilization , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish , Zebrafish Proteins/genetics , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics , beta-Synuclein/genetics
5.
Biochemistry ; 50(34): 7295-310, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21797260

ABSTRACT

Plasma membrane and vesicular transporters for the biogenic amines, dopamine, norepinephrine, and serotonin, represent a group of proteins that play a crucial role in the regulation of neurotransmission. Clinically, mono amine transporters are the primary targets for the actions of many therapeutic agents used to treat mood disorders, as well as the site of action for highly addictive psychostimulants such as cocaine, amphetamine, methamphetamine, and 3,4-methylenedioxymethamphetamine. Over the past decade, the use of approaches such as yeast two-hybrid and proteomics has identified a multitude of transporter interacting proteins, suggesting that the function and regulation of these transporters are more complex than previously anticipated. With the increasing number of interacting proteins, the rules dictating transporter synthesis, assembly, targeting, trafficking, and function are beginning to be deciphered. Although many of these protein interactions have yet to be fully characterized, current knowledge is beginning to shed light on novel transporter mechanisms involved in monoamine homeostasis, the molecular actions of psychostimulants, and potential disease mechanisms. While future studies resolving the spatial and temporal resolution of these, and yet unknown, interactions will be needed, the realization that monoamine transporters do not work alone opens the path to a plethora of possible pharmacological interventions.


Subject(s)
Vesicular Monoamine Transport Proteins/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Humans , Protein Binding , Protein Transport , Transport Vesicles/metabolism , Vesicular Monoamine Transport Proteins/biosynthesis
6.
Brain Struct Funct ; 214(2-3): 285-302, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20162303

ABSTRACT

Since the introduction of the zebrafish as a model for the study of vertebrate developmental biology, an extensive array of techniques for its experimental manipulation and analysis has been developed. Recently it has become apparent that these powerful methodologies might be deployed in order to elucidate the pathogenesis of human neurodegenerative diseases and to identify candidate therapeutic approaches. In this article, we consider evidence that the zebrafish central nervous system provides an appropriate setting in which to model human neurological disease and we review techniques and resources available for generating transgenic models. We then examine recent publications showing that appropriate phenotypes can be provoked in the zebrafish through transgenic manipulations analogous to genetic abnormalities known to cause human tauopathies, polyglutamine diseases or motor neuron degenerations. These studies show proof of concept that findings in zebrafish models can be applicable to the pathogenic mechanisms underlying human diseases. Consequently, the prospects for providing novel insights into neurodegenerative diseases by exploiting transgenic zebrafish models and discovery-driven approaches seem favorable.


Subject(s)
Animals, Genetically Modified/genetics , Molecular Biology/methods , Neurodegenerative Diseases/genetics , Zebrafish/genetics , Animals , Disease Models, Animal , Genetic Predisposition to Disease/genetics , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Molecular Biology/trends , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , Motor Neuron Disease/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Phenotype , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL