Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
PLoS One ; 19(3): e0300529, 2024.
Article in English | MEDLINE | ID: mdl-38498506

ABSTRACT

Behavioral thresholds define the lowest stimulus intensities sufficient to elicit a behavioral response. Establishment of baseline behavioral thresholds during development is critical for proper responses throughout the animal's life. Despite the relevance of such innate thresholds, the molecular mechanisms critical to establishing behavioral thresholds during development are not well understood. The acoustic startle response is a conserved behavior whose threshold is established during development yet is subsequently acutely regulated. We have previously identified a zebrafish mutant line (escapist) that displays a decreased baseline or innate acoustic startle threshold. Here, we identify a single base pair substitution on Chromosome 25 located within the coding sequence of the synaptotagmin 7a (syt7a) gene that is tightly linked to the escapist acoustic hypersensitivity phenotype. By generating animals in which we deleted the syt7a open reading frame, and subsequent complementation testing with the escapist line, we demonstrate that loss of syt7a function is not the cause of the escapist behavioral phenotype. Nonetheless, escapist mutants provide a powerful tool to decipher the overlap between acute and developmental regulation of behavioral thresholds. Extensive behavioral analyses reveal that in escapist mutants the establishment of the innate acoustic startle threshold is impaired, while regulation of its acute threshold remains intact. Moreover, our behavioral analyses reveal a deficit in baseline responses to visual stimuli, but not in the acute regulation of responses to visual stimuli. Together, this work eliminates loss of syt7a as causative for the escapist phenotype and suggests that mechanisms that regulate the establishment of behavioral thresholds in escapist larvae can operate independently from those regulating acute threshold regulation.


Subject(s)
Reflex, Startle , Zebrafish , Animals , Reflex, Startle/genetics , Zebrafish/genetics , Base Pairing , Acoustic Stimulation , Behavior, Animal/physiology
2.
bioRxiv ; 2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37662318

ABSTRACT

Behavioral thresholds define the lowest stimulus intensities sufficient to elicit a behavioral response. Establishment of baseline behavioral thresholds during development is critical for proper responses throughout the animal's life. Despite the relevance of such innate thresholds, the molecular mechanisms critical to establishing behavioral thresholds during development are not well understood. The acoustic startle response is a conserved behavior whose threshold is established during development yet is subsequently acutely regulated. We have previously identified a zebrafish mutant line ( escapist ) that displays a decreased baseline or innate acoustic startle threshold. Here, we identify a single base pair substitution on Chromosome 25 located within the coding sequence of the synaptotagmin 7a ( syt7a ) gene that is tightly linked to the escapist acoustic hypersensitivity phenotype. By generating animals in which we deleted the syt7a open reading frame, and subsequent complementation testing with the escapist line, we demonstrate that loss of syt7a function is not the cause of the escapist behavioral phenotype. Nonetheless, escapist mutants provide a powerful tool to decipher the overlap between acute and developmental regulation of behavioral thresholds. Extensive behavioral analyses reveal that in escapist mutants the establishment of the innate acoustic startle threshold is impaired, while regulation of its acute threshold remains intact. Moreover, our behavioral analyses reveal a deficit in baseline responses to visual stimuli, but not in the acute regulation of responses to visual stimuli. Together, this work eliminates loss of syt7a as causative for the escapist phenotype and suggests that mechanisms that regulate the establishment of behavioral thresholds in escapist larvae can operate largely independently from those regulating acute threshold regulation.

3.
PLoS Genet ; 19(3): e1010650, 2023 03.
Article in English | MEDLINE | ID: mdl-36972301

ABSTRACT

Habituation is a foundational learning process critical for animals to adapt their behavior to changes in their sensory environment. Although habituation is considered a simple form of learning, the identification of a multitude of molecular pathways including several neurotransmitter systems that regulate this process suggests an unexpected level of complexity. How the vertebrate brain integrates these various pathways to accomplish habituation learning, whether they act independently or intersect with one another, and whether they act via divergent or overlapping neural circuits has remained unclear. To address these questions, we combined pharmacogenetic pathway analysis with unbiased whole-brain activity mapping using the larval zebrafish. Based on our findings, we propose five distinct molecular modules for the regulation of habituation learning and identify a set of molecularly defined brain regions associated with four of the five modules. Moreover, we find that in module 1 the palmitoyltransferase Hip14 cooperates with dopamine and NMDA signaling to drive habituation, while in module 3 the adaptor protein complex subunit Ap2s1 drives habituation by antagonizing dopamine signaling, revealing two distinct and opposing roles for dopaminergic neuromodulation in the regulation of behavioral plasticity. Combined, our results define a core set of distinct modules that we propose act in concert to regulate habituation-associated plasticity, and provide compelling evidence that even seemingly simple learning behaviors in a compact vertebrate brain are regulated by a complex and overlapping set of molecular mechanisms.


Subject(s)
Habituation, Psychophysiologic , Zebrafish , Animals , Zebrafish/genetics , Habituation, Psychophysiologic/physiology , Dopamine , Learning/physiology , Brain , Neuronal Plasticity/genetics
4.
PLoS One ; 17(7): e0270903, 2022.
Article in English | MEDLINE | ID: mdl-35834485

ABSTRACT

BACKGROUND: The ability to filter sensory information into relevant versus irrelevant stimuli is a fundamental, conserved property of the central nervous system and is accomplished in part through habituation learning. Synaptic plasticity that underlies habituation learning has been described at the cellular level, yet the genetic regulators of this plasticity remain poorly understood, as do circuits that mediate sensory filtering. METHODS: To identify genes critical for plasticity, a forward genetic screen for zebrafish genes that mediate habituation learning was performed, which identified a mutant allele, doryp177, that caused reduced habituation of the acoustic startle response. In this study, we combine whole-genome sequencing with behavioral analyses to characterize and identify the gene affected in doryp177 mutants. RESULTS: Whole-genome sequencing identified the calcium voltage-gated channel auxiliary subunit alpha-2/delta-3 (cacna2d3) as a candidate gene affected in doryp177 mutants. Behavioral characterization of larvae homozygous for two additional, independently derived mutant alleles of cacna2d3, together with failure of these alleles to complement doryp177, confirmed a critical role for cacna2d3 in habituation learning. Notably, detailed analyses of the acoustic response in mutant larvae also revealed increased startle sensitivity to acoustic stimuli, suggesting a broader role for cacna2d3 in controlling innate response thresholds to acoustic stimuli. CONCLUSIONS: Taken together, our data demonstrate a critical role for cacna2d3 in sensory filtering, a process that is disrupted in human CNS disorders, e.g. ADHD, schizophrenia, and autism.


Subject(s)
Calcium Channels , Habituation, Psychophysiologic , Reflex, Startle , Zebrafish , Acoustic Stimulation , Animals , Calcium Channels/genetics , Habituation, Psychophysiologic/genetics , Larva/genetics , Learning/physiology , Reflex, Startle/genetics , Zebrafish/genetics
5.
Development ; 149(9)2022 05 01.
Article in English | MEDLINE | ID: mdl-35552393

ABSTRACT

Nervous system assembly relies on a diversity of cellular processes ranging from dramatic tissue reorganization to local, subcellular changes all driven by precise molecular programs. Combined, these processes culminate in an animal's ability to plan and execute behaviors. Animal behavior can, therefore, serve as a functional readout of nervous system development. Benefitting from an expansive and growing set of molecular and imaging tools paired with an ever-growing number of assays of diverse behaviors, the zebrafish system has emerged as an outstanding platform at the intersection of nervous system assembly, plasticity and behavior. Here, we summarize recent advancements in the field, including how developing neural circuits are refined to shape complex behaviors and plasticity.


Subject(s)
Nervous System , Zebrafish , Animals , Behavior, Animal/physiology , Neuronal Plasticity/physiology
6.
PLoS Genet ; 17(6): e1008943, 2021 06.
Article in English | MEDLINE | ID: mdl-34061829

ABSTRACT

The acoustic startle response is an evolutionarily conserved avoidance behavior. Disruptions in startle behavior, particularly startle magnitude, are a hallmark of several human neurological disorders. While the neural circuitry underlying startle behavior has been studied extensively, the repertoire of genes and genetic pathways that regulate this locomotor behavior has not been explored using an unbiased genetic approach. To identify such genes, we took advantage of the stereotypic startle behavior in zebrafish larvae and performed a forward genetic screen coupled with whole genome analysis. We uncovered mutations in eight genes critical for startle behavior, including two genes encoding proteins associated with human neurological disorders, Dolichol kinase (Dolk), a broadly expressed regulator of the glycoprotein biosynthesis pathway, and the potassium Shaker-like channel subunit Kv1.1. We demonstrate that Kv1.1 and Dolk play critical roles in the spinal cord to regulate movement magnitude during the startle response and spontaneous swim movements. Moreover, we show that Kv1.1 protein is mislocalized in dolk mutants, suggesting they act in a common genetic pathway. Combined, our results identify a diverse set of eight genes, all associated with human disorders, that regulate zebrafish startle behavior and reveal a previously unappreciated role for Dolk and Kv1.1 in regulating movement magnitude via a common genetic pathway.


Subject(s)
Genetic Testing/methods , Kv1.1 Potassium Channel/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Reflex, Startle/genetics , Zebrafish Proteins/genetics , Animals , Humans , Phosphotransferases (Alcohol Group Acceptor)/genetics , Zebrafish
7.
Curr Biol ; 30(14): 2729-2738.e4, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32502414

ABSTRACT

Habituation is an adaptive learning process that enables animals to adjust innate behaviors to changes in their environment. Despite its well-documented implications for a wide diversity of behaviors, the molecular and cellular basis of habituation learning is not well understood. Using whole-genome sequencing of zebrafish mutants isolated in an unbiased genetic screen, we identified the palmitoyltransferase Huntingtin interacting protein 14 (Hip14) as a critical regulator of habituation learning. We demonstrate that Hip14 regulates depression of sensory inputs onto an identified hindbrain neuron and provide evidence that Hip14 palmitoylates the Shaker-like K+ voltage-gated channel subunit (Kv1.1), thereby regulating Kv1.1 subcellular localization. Furthermore, we show that, like for Hip14, loss of Kv1.1 leads to habituation deficits and that Hip14 is dispensable in development and instead acts acutely to promote habituation. Combined, these results uncover a previously unappreciated role for acute posttranslational palmitoylation at defined circuit components to regulate learning.


Subject(s)
Acyltransferases/physiology , Adaptor Proteins, Signal Transducing/physiology , Habituation, Psychophysiologic/genetics , Learning/physiology , Lipoylation/genetics , Lipoylation/physiology , Nerve Tissue Proteins/physiology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Shaker Superfamily of Potassium Channels/physiology , Zebrafish/genetics , Zebrafish/physiology , Animals , Presynaptic Terminals/metabolism , Shaker Superfamily of Potassium Channels/metabolism
8.
Cell ; 176(1-2): 85-97.e14, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30580965

ABSTRACT

Animals must respond to the ingestion of food by generating adaptive behaviors, but the role of gut-brain signaling in behavioral regulation is poorly understood. Here, we identify conserved ion channels in an enteric serotonergic neuron that mediate its responses to food ingestion and decipher how these responses drive changes in foraging behavior. We show that the C. elegans serotonergic neuron NSM acts as an enteric sensory neuron that acutely detects food ingestion. We identify the novel and conserved acid-sensing ion channels (ASICs) DEL-7 and DEL-3 as NSM-enriched channels required for feeding-dependent NSM activity, which in turn drives slow locomotion while animals feed. Point mutations that alter the DEL-7 channel change NSM dynamics and associated behavioral dynamics of the organism. This study provides causal links between food ingestion, molecular and physiological properties of an enteric serotonergic neuron, and adaptive feeding behaviors, yielding a new view of how enteric neurons control behavior.


Subject(s)
Acid Sensing Ion Channels/metabolism , Enteric Nervous System/metabolism , Feeding Behavior/physiology , Acid Sensing Ion Channels/physiology , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Enteric Nervous System/physiology , Food , Ion Channels/metabolism , Ion Channels/physiology , Locomotion , Neurons/metabolism , Sensory Receptor Cells/metabolism , Serotonergic Neurons/metabolism , Serotonergic Neurons/physiology , Serotonin , Signal Transduction
9.
Cell Rep ; 23(3): 878-887, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669291

ABSTRACT

Sensory experiences dynamically modify whether animals respond to a given stimulus, but it is unclear how innate behavioral thresholds are established. Here, we identify molecular and circuit-level mechanisms underlying the innate threshold of the zebrafish startle response. From a forward genetic screen, we isolated five mutant lines with reduced innate startle thresholds. Using whole-genome sequencing, we identify the causative mutation for one line to be in the fragile X mental retardation protein (FMRP)-interacting protein cyfip2. We show that cyfip2 acts independently of FMRP and that reactivation of cyfip2 restores the baseline threshold after phenotype onset. Finally, we show that cyfip2 regulates the innate startle threshold by reducing neural activity in a small group of excitatory hindbrain interneurons. Thus, we identify a selective set of genes critical to establishing an innate behavioral threshold and uncover a circuit-level role for cyfip2 in this process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Interneurons/metabolism , Zebrafish Proteins/metabolism , Acoustic Stimulation , Adaptor Proteins, Signal Transducing/genetics , Animals , Axons/metabolism , Behavior, Animal , Calcium/metabolism , Cytoskeleton/metabolism , Excitatory Postsynaptic Potentials , Fragile X Mental Retardation Protein/metabolism , Hypersensitivity/metabolism , Hypersensitivity/pathology , Larva/metabolism , Mutagenesis , Reflex, Startle/physiology , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics
10.
Curr Biol ; 28(9): 1357-1369.e5, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29681477

ABSTRACT

Animals continuously integrate sensory information and select contextually appropriate responses. Here, we show that zebrafish larvae select a behavioral response to acoustic stimuli from a pre-existing choice repertoire in a context-dependent manner. We demonstrate that this sensorimotor choice is modulated by stimulus quality and history, as well as by neuromodulatory systems-all hallmarks of more complex decision making. Moreover, from a genetic screen coupled with whole-genome sequencing, we identified eight mutants with deficits in this sensorimotor choice, including mutants of the vertebrate-specific G-protein-coupled extracellular calcium-sensing receptor (CaSR), whose function in the nervous system is not well understood. We demonstrate that CaSR promotes sensorimotor decision making acutely through Gαi/o and Gαq/11 signaling, modulated by clathrin-mediated endocytosis. Combined, our results identify the first set of genes critical for behavioral choice modulation in a vertebrate and reveal an unexpected critical role for CaSR in sensorimotor decision making.


Subject(s)
Choice Behavior/physiology , Mutation , Psychomotor Performance , Receptors, Calcium-Sensing/physiology , Zebrafish Proteins/physiology , Zebrafish/physiology , Acoustic Stimulation , Animals , Behavior, Animal , Calcium/metabolism , Genetic Testing , Receptors, Calcium-Sensing/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics
11.
Neuron ; 90(2): 278-91, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27068791

ABSTRACT

Changes in neuronal activity create local and transient changes in energy demands at synapses. Here we discover a metabolic compartment that forms in vivo near synapses to meet local energy demands and support synaptic function in Caenorhabditis elegans neurons. Under conditions of energy stress, glycolytic enzymes redistribute from a diffuse localization in the cytoplasm to a punctate localization adjacent to synapses. Glycolytic enzymes colocalize, suggesting the ad hoc formation of a glycolysis compartment, or a "glycolytic metabolon," that can maintain local levels of ATP. Local formation of the glycolytic metabolon is dependent on presynaptic scaffolding proteins, and disruption of the glycolytic metabolon blocks the synaptic vesicle cycle, impairs synaptic recovery, and affects locomotion. Our studies indicate that under energy stress conditions, energy demands in C. elegans synapses are met locally through the assembly of a glycolytic metabolon to sustain synaptic function and behavior. VIDEO ABSTRACT.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Phosphofructokinase-1/metabolism , Presynaptic Terminals/enzymology , Presynaptic Terminals/physiology , Stress, Physiological , Animals , Caenorhabditis elegans/metabolism , Endocytosis , Hypoxia , Metabolomics , Mutation , Presynaptic Terminals/metabolism , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism
12.
Development ; 141(2): 422-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353061

ABSTRACT

Transcription factors that drive neuron type-specific terminal differentiation programs in the developing nervous system are often expressed in several distinct neuronal cell types, but to what extent they have similar or distinct activities in individual neuronal cell types is generally not well explored. We investigate this problem using, as a starting point, the C. elegans LIM homeodomain transcription factor ttx-3, which acts as a terminal selector to drive the terminal differentiation program of the cholinergic AIY interneuron class. Using a panel of different terminal differentiation markers, including neurotransmitter synthesizing enzymes, neurotransmitter receptors and neuropeptides, we show that ttx-3 also controls the terminal differentiation program of two additional, distinct neuron types, namely the cholinergic AIA interneurons and the serotonergic NSM neurons. We show that the type of differentiation program that is controlled by ttx-3 in different neuron types is specified by a distinct set of collaborating transcription factors. One of the collaborating transcription factors is the POU homeobox gene unc-86, which collaborates with ttx-3 to determine the identity of the serotonergic NSM neurons. unc-86 in turn operates independently of ttx-3 in the anterior ganglion where it collaborates with the ARID-type transcription factor cfi-1 to determine the cholinergic identity of the IL2 sensory and URA motor neurons. In conclusion, transcription factors operate as terminal selectors in distinct combinations in different neuron types, defining neuron type-specific identity features.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Homeodomain Proteins/genetics , Neurons/cytology , Neurons/metabolism , Neuropeptides/genetics , POU Domain Factors/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Differentiation/genetics , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Gene Expression Regulation, Developmental , Genes, Helminth , Homeodomain Proteins/metabolism , Interneurons/cytology , Interneurons/metabolism , Larva/cytology , Larva/growth & development , Larva/metabolism , Neurogenesis/genetics , Neurons/classification , Neuropeptides/metabolism , POU Domain Factors/metabolism , Serotonergic Neurons/cytology , Serotonergic Neurons/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cell Adh Migr ; 7(4): 379-87, 2013.
Article in English | MEDLINE | ID: mdl-23628914

ABSTRACT

Dramatic morphogenetic processes underpin nearly every step of nervous system development, from initial neuronal migration and axon guidance to synaptogenesis. Underlying this morphogenesis are dynamic rearrangements of cytoskeletal architecture. Here we discuss the roles of the actin cytoskeleton in the development of presynaptic terminals, from the elaboration of terminal arbors to the recruitment of presynaptic vesicles and active zone components. The studies discussed here underscore the importance of actin regulation at every step in neuronal circuit assembly.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Synapses/metabolism , Animals , Models, Biological , Synaptic Vesicles/metabolism
14.
J Neurosci ; 33(4): 1366-76, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23345213

ABSTRACT

Neurosecretory release sites lack distinct postsynaptic partners, yet target to specific circuits. This targeting specificity regulates local release of neurotransmitters and modulation of adjacent circuits. How neurosecretory release sites target to specific regions is not understood. Here we identify a molecular mechanism that governs the spatial specificity of extrasynaptic neurosecretory terminal (ENT) formation in the serotonergic neurosecretory-motor (NSM) neurons of Caenorhabditis elegans. We show that postembryonic arborization and neurosecretory terminal targeting of the C. elegans NSM neuron is dependent on the Netrin receptor UNC-40/DCC. We observe that UNC-40 localizes to specific neurosecretory terminals at the time of axon arbor formation. This localization is dependent on UNC-6/Netrin, which is expressed by nerve ring neurons that act as guideposts to instruct local arbor and release site formation. We find that both UNC-34/Enabled and MIG-10/Lamellipodin are required downstream of UNC-40 to link the sites of ENT formation to nascent axon arbor extensions. Our findings provide a molecular link between release site development and axon arborization and introduce a novel mechanism that governs the spatial specificity of serotonergic ENTs in vivo.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serotonergic Neurons/physiology , Synaptic Transmission/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/embryology , Cell Communication/physiology , Image Processing, Computer-Assisted , Microscopy, Confocal , Netrins , Neurons/metabolism , Neurons/ultrastructure , Serotonergic Neurons/ultrastructure , Synapses/physiology , Synapses/ultrastructure
15.
Psychol Sci ; 24(2): 213-8, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23232861

ABSTRACT

This study used a signal detection paradigm to explore the Marley hypothesis--that group differences in perception of racism reflect dominant-group denial of and ignorance about the extent of past racism. White American students from a midwestern university and Black American students from two historically Black universities completed surveys about their historical knowledge and perception of racism. Relative to Black participants, White participants perceived less racism in both isolated incidents and systemic manifestations of racism. They also performed worse on a measure of historical knowledge (i.e., they did not discriminate historical fact from fiction), and this group difference in historical knowledge mediated the differences in perception of racism. Racial identity relevance moderated group differences in perception of systemic manifestations of racism (but not isolated incidents), such that group differences were stronger among participants who scored higher on a measure of racial identity relevance. The results help illuminate the importance of epistemologies of ignorance: cultural-psychological tools that afford denial of and inaction about injustice.


Subject(s)
Black or African American/psychology , Denial, Psychological , History , Racism/psychology , Social Perception , White People/psychology , Humans
16.
Genes Dev ; 26(19): 2206-21, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23028145

ABSTRACT

The chemotrophic factor Netrin can simultaneously instruct different neurodevelopmental programs in individual neurons in vivo. How neurons correctly interpret the Netrin signal and undergo the appropriate neurodevelopmental response is not understood. Here we identify MIG-10 isoforms as critical determinants of individual cellular responses to Netrin. We determined that distinct MIG-10 isoforms, varying only in their N-terminal motifs, can localize to specific subcellular domains and are differentially required for discrete neurodevelopmental processes in vivo. We identified MIG-10B as an isoform uniquely capable of localizing to presynaptic regions and instructing synaptic vesicle clustering in response to Netrin. MIG-10B interacts with Abl-interacting protein-1 (ABI-1)/Abi1, a component of the WAVE complex, to organize the actin cytoskeleton at presynaptic sites and instruct vesicle clustering through SNN-1/Synapsin. We identified a motif in the MIG-10B N-terminal domain that is required for its function and localization to presynaptic sites. With this motif, we engineered a dominant-negative MIG-10B construct that disrupts vesicle clustering and animal thermotaxis behavior when expressed in a single neuron in vivo. Our findings indicate that the unique N-terminal domains confer distinct MIG-10 isoforms with unique capabilities to localize to distinct subcellular compartments, organize the actin cytoskeleton at these sites, and instruct distinct Netrin-dependent neurodevelopmental programs.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/genetics , Synaptic Vesicles/metabolism , Actin Cytoskeleton/metabolism , Animals , Behavior, Animal/physiology , Caenorhabditis elegans/metabolism , Cell Movement , Cytoskeletal Proteins/genetics , Gene Expression Profiling , Interneurons/cytology , Motor Neurons/cytology , Nerve Tissue Proteins/metabolism , Netrins , Protein Isoforms , Protein Transport/genetics , Synaptic Vesicles/genetics
17.
Biophys J ; 100(12): L63-5, 2011 Jun 22.
Article in English | MEDLINE | ID: mdl-21689517

ABSTRACT

We report superresolution fluorescence microscopy in an intact living organism, namely Caenorhabditis elegans nematodes expressing green fluorescent protein (GFP)-fusion proteins. We also superresolve, by stimulated emission depletion (STED) microscopy, living cultured cells, demonstrating that STED microscopy with GFP can be widely applied. STED with GFP can be performed with both pulsed and continuous-wave lasers spanning a wide wavelength range from at least 556-592 nm. Acquiring subdiffraction resolution images within seconds enables the recording of movies revealing structural dynamics. These results demonstrate that numerous microscopy studies of live samples employing GFP as the marker can be performed at subdiffraction resolution.


Subject(s)
Caenorhabditis elegans/metabolism , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence/methods , Nanotechnology/methods , Saccharomyces cerevisiae/metabolism , Absorption , Animals , Caenorhabditis elegans/cytology , Neurons/cytology , Neurons/metabolism , Saccharomyces cerevisiae/cytology , Spectrum Analysis
18.
Microbiology (Reading) ; 154(Pt 8): 2231-2240, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18667556

ABSTRACT

Adaptation to the acidic microenvironment, and adherence to mucosal epithelium, are essential for persistent colonization of the human stomach by Helicobacter pylori. The expression of SabA, an adhesin implicated in the ability of H. pylori to adhere to the host gastric epithelium, can be modulated by phase variation via slipped-strand mispairing in repetitive nucleotide tracts located in both the promoter region and the coding region. This study demonstrates the occurrence of phase variation at the sabA locus within individual strains of H. pylori, and among multiple isolates from a single patient. In addition, transcription of sabA is repressed by the acid-responsive ArsRS two-component signal transduction system in vitro. Our results demonstrate that isogenic inactivation of the arsS (jhp0151/HP0165) histidine kinase locus results in a 10-fold SabA-dependent increase in adherence to gastric epithelial cells in strain J99 (contains an in-frame sabA allele), but not in strain 26695 (out-of-frame sabA allele). The combination of transcriptional regulation of the sabA locus by the ArsRS two-component signal-transduction system and the generation of subpopulations harbouring alternate sabA alleles by slipped-strand mispairing during chromosomal replication could permit H. pylori to rapidly adapt to varying microenvironments or host immune responses. As a pathogen with a paucity of regulatory proteins, this dual regulation indicates that SabA expression is a tightly regulated process in H. pylori infection.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Protein Kinases/metabolism , Signal Transduction , Adhesins, Bacterial/genetics , Bacterial Adhesion , Bacterial Proteins/genetics , Cell Line, Tumor , Gastric Mucosa/microbiology , Helicobacter pylori/genetics , Histidine Kinase , Humans , Phenotype , Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...