Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Cell Sci ; 137(7)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38477340

RESUMO

Axonal transport in neurons is essential for cargo movement between the cell body and synapses. Caenorhabditis elegans UNC-104 and its homolog KIF1A are kinesin-3 motors that anterogradely transport precursors of synaptic vesicles (pre-SVs) and are degraded at synapses. However, in C. elegans, touch neuron-specific knockdown of the E1 ubiquitin-activating enzyme, uba-1, leads to UNC-104 accumulation at neuronal ends and synapses. Here, we performed an RNAi screen and identified that depletion of fbxb-65, which encodes an F-box protein, leads to UNC-104 accumulation at neuronal distal ends, and alters UNC-104 net anterograde movement and levels of UNC-104 on cargo without changing synaptic UNC-104 levels. Split fluorescence reconstitution showed that UNC-104 and FBXB-65 interact throughout the neuron. Our theoretical model suggests that UNC-104 might exhibit cooperative cargo binding that is regulated by FBXB-65. FBXB-65 regulates an unidentified post-translational modification (PTM) of UNC-104 in a region beside the cargo-binding PH domain. Both fbxb-65 and UNC-104, independently of FBXB-65, regulate axonal pre-SV distribution, transport of pre-SVs at branch points and organismal lifespan. FBXB-65 regulates a PTM of UNC-104 and the number of motors on the cargo surface, which can fine-tune cargo transport to the synapse.


Assuntos
Transporte Axonal , Proteínas de Caenorhabditis elegans , Proteínas F-Box , Cinesinas , Animais , Transporte Axonal/fisiologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas F-Box/metabolismo , Cinesinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Domínios de Homologia à Plecstrina , Processamento de Proteína Pós-Traducional
2.
PLoS Genet ; 19(11): e1011005, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37934770

RESUMO

BACKGROUND: Kinesin motor proteins transport intracellular cargo, including mRNA, proteins, and organelles. Pathogenic variants in kinesin-related genes have been implicated in neurodevelopmental disorders and skeletal dysplasias. We identified de novo, heterozygous variants in KIF5B, encoding a kinesin-1 subunit, in four individuals with osteogenesis imperfecta. The variants cluster within the highly conserved kinesin motor domain and are predicted to interfere with nucleotide binding, although the mechanistic consequences on cell signaling and function are unknown. METHODS: To understand the in vivo genetic mechanism of KIF5B variants, we modeled the p.Thr87Ile variant that was found in two patients in the C. elegans ortholog, unc-116, at the corresponding position (Thr90Ile) by CRISPR/Cas9 editing and performed functional analysis. Next, we studied the cellular and molecular consequences of the recurrent p.Thr87Ile variant by microscopy, RNA and protein analysis in NIH3T3 cells, primary human fibroblasts and bone biopsy. RESULTS: C. elegans heterozygous for the unc-116 Thr90Ile variant displayed abnormal body length and motility phenotypes that were suppressed by additional copies of the wild type allele, consistent with a dominant negative mechanism. Time-lapse imaging of GFP-tagged mitochondria showed defective mitochondria transport in unc-116 Thr90Ile neurons providing strong evidence for disrupted kinesin motor function. Microscopy studies in human cells showed dilated endoplasmic reticulum, multiple intracellular vacuoles, and abnormal distribution of the Golgi complex, supporting an intracellular trafficking defect. RNA sequencing, proteomic analysis, and bone immunohistochemistry demonstrated down regulation of the mTOR signaling pathway that was partially rescued with leucine supplementation in patient cells. CONCLUSION: We report dominant negative variants in the KIF5B kinesin motor domain in individuals with osteogenesis imperfecta. This study expands the spectrum of kinesin-related disorders and identifies dysregulated signaling targets for KIF5B in skeletal development.


Assuntos
Cinesinas , Osteogênese Imperfeita , Animais , Humanos , Camundongos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Transporte/genética , Regulação para Baixo , Cinesinas/genética , Cinesinas/metabolismo , Células NIH 3T3 , Proteômica , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
3.
Traffic ; 19(3): 166-181, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29178177

RESUMO

Steady axonal cargo flow is central to the functioning of healthy neurons. However, a substantial fraction of cargo in axons remains stationary up to several minutes. We examine the transport of precursors of synaptic vesicles (pre-SVs), endosomes and mitochondria in Caenorhabditis elegans touch receptor neurons, showing that stationary cargo are predominantly present at actin-rich regions along the neuronal process. Stationary vesicles at actin-rich regions increase the propensity of moving vesicles to stall at the same location, resulting in traffic jams arising from physical crowding. Such local traffic jams at actin-rich regions are likely to be a general feature of axonal transport since they also occur in Drosophila neurons. Repeated touch stimulation of C. elegans reduces the density of stationary pre-SVs, indicating that these traffic jams can act as both sources and sinks of vesicles. This suggests that vesicles trapped in actin-rich regions are functional reservoirs that may contribute to maintaining robust cargo flow in the neuron. A video abstract of this article can be found at: Video S1; Video S2.


Assuntos
Transporte Axonal , Actinas/metabolismo , Animais , Caenorhabditis elegans , Drosophila , Endossomos/metabolismo , Mitocôndrias/metabolismo , Vesículas Sinápticas/metabolismo
4.
Development ; 141(20): 3922-33, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25252943

RESUMO

We describe the identification of zyxin as a regulator of synapse maintenance in mechanosensory neurons in C. elegans. zyx-1 mutants lacked PLM mechanosensory synapses as adult animals. However, most PLM synapses initially formed during development but were subsequently lost as the animals developed. Vertebrate zyxin regulates cytoskeletal responses to mechanical stress in culture. Our work provides in vivo evidence in support of such a role for zyxin. In particular, zyx-1 mutant synaptogenesis phenotypes were suppressed by disrupting locomotion of the mutant animals, suggesting that zyx-1 protects mechanosensory synapses from locomotion-induced forces. In cultured cells, zyxin is recruited to focal adhesions and stress fibers via C-terminal LIM domains and modulates cytoskeletal organization via the N-terminal domain. The synapse-stabilizing activity was mediated by a short isoform of ZYX-1 containing only the LIM domains. Consistent with this notion, PLM synaptogenesis was independent of α-actinin and ENA-VASP, both of which bind to the N-terminal domain of zyxin. Our results demonstrate that the LIM domain moiety of zyxin functions autonomously to mediate responses to mechanical stress and provide in vivo evidence for a role of zyxin in neuronal development.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Sinapses/fisiologia , Zixina/fisiologia , Actinina/metabolismo , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Proteínas de Transporte/química , Citoesqueleto/metabolismo , Adesões Focais/metabolismo , Movimento , Mutação , Neurônios/metabolismo , Fenótipo , Fosfoproteínas/química , Isoformas de Proteínas/fisiologia , Estrutura Terciária de Proteína , Estresse Mecânico
5.
PLoS Genet ; 10(10): e1004644, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25329901

RESUMO

Axonal transport of synaptic vesicles (SVs) is a KIF1A/UNC-104 mediated process critical for synapse development and maintenance yet little is known of how SV transport is regulated. Using C. elegans as an in vivo model, we identified SAM-4 as a novel conserved vesicular component regulating SV transport. Processivity, but not velocity, of SV transport was reduced in sam-4 mutants. sam-4 displayed strong genetic interactions with mutations in the cargo binding but not the motor domain of unc-104. Gain-of-function mutations in the unc-104 motor domain, identified in this study, suppress the sam-4 defects by increasing processivity of the SV transport. Genetic analyses suggest that SAM-4, SYD-2/liprin-α and the KIF1A/UNC-104 motor function in the same pathway to regulate SV transport. Our data support a model in which the SV protein SAM-4 regulates the processivity of SV transport.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Membrana/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal/genética , Sítios de Ligação , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas de Membrana/genética , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Neurônios/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo
6.
Nucleic Acids Res ; 42(22): 13778-87, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25399416

RESUMO

To study target sequence specificity, selectivity, and reaction kinetics of Streptococcus pyogenes Cas9 activity, we challenged libraries of random variant targets with purified Cas9::guide RNA complexes in vitro. Cleavage kinetics were nonlinear, with a burst of initial activity followed by slower sustained cleavage. Consistent with other recent analyses of Cas9 sequence specificity, we observe considerable (albeit incomplete) impairment of cleavage for targets mutated in the PAM sequence or in 'seed' sequences matching the proximal 8 bp of the guide. A second target region requiring close homology was located at the other end of the guide::target duplex (positions 13-18 relative to the PAM). Sequences flanking the guide+PAM region had measurable (albeit modest) effects on cleavage. In addition, the first-base Guanine constraint commonly imposed by gRNA expression systems has little effect on overall cleavage efficiency. Taken together, these studies provide an in vitro understanding of the complexities of Cas9-gRNA interaction and cleavage beyond the general paradigm of site determination based on the 'seed' sequence and PAM.


Assuntos
Proteínas Associadas a CRISPR/metabolismo , Clivagem do DNA , Endodesoxirribonucleases/metabolismo , DNA/química , Cinética , RNA/química , Homologia de Sequência do Ácido Nucleico , Streptococcus pyogenes/enzimologia
7.
Development ; 138(1): 87-96, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21115607

RESUMO

Little is known about transcriptional control of neurite branching or presynaptic differentiation, events that occur relatively late in neuronal development. Using the Caenorhabditis elegans mechanosensory circuit as an in vivo model, we show that SAM-10, an ortholog of mammalian single-stranded DNA-binding protein (SSDP), functions cell-autonomously in the nucleus to regulate synaptic differentiation, as well as positioning of, a single neurite branch. PLM mechanosensory neurons in sam-10 mutants exhibit abnormal placement of the neurite branch point, and defective synaptogenesis, characterized by an overextended synaptic varicosity, underdeveloped synaptic morphology and disrupted colocalization of active zone and synaptic vesicles. SAM-10 functions coordinately with Lim domain-binding protein 1 (LDB-1), demonstrated by our observations that: (1) mutations in either gene show similar defects in PLM neurons; and (2) LDB-1 is required for SAM-10 nuclear localization. SAM-10 regulates PLM synaptic differentiation by suppressing transcription of prk-2, which encodes an ortholog of the mammalian Pim kinase family. PRK-2-mediated activities of SAM-10 are specifically involved in PLM synaptic differentiation, but not other sam-10 phenotypes such as neurite branching. Thus, these data reveal a novel transcriptional signaling pathway that regulates neuronal specification of neurite branching and presynaptic differentiation.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriologia , Diferenciação Celular/fisiologia , Neuritos/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Microscopia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/metabolismo
8.
PLoS Genet ; 6(11): e1001200, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21079789

RESUMO

UNC-104/KIF1A is a Kinesin-3 motor that transports synaptic vesicles from the cell body towards the synapse by binding to PI(4,5)P(2) through its PH domain. The fate of the motor upon reaching the synapse is not known. We found that wild-type UNC-104 is degraded at synaptic regions through the ubiquitin pathway and is not retrogradely transported back to the cell body. As a possible means to regulate the motor, we tested the effect of cargo binding on UNC-104 levels. The unc-104(e1265) allele carries a point mutation (D1497N) in the PI(4,5)P(2) binding pocket of the PH domain, resulting in greatly reduced preferential binding to PI(4,5)P(2)in vitro and presence of very few motors on pre-synaptic vesicles in vivo. unc-104(e1265) animals have poor locomotion irrespective of in vivo PI(4,5)P(2) levels due to reduced anterograde transport. Moreover, they show highly reduced levels of UNC-104 in vivo. To confirm that loss of cargo binding specificity reduces motor levels, we isolated two intragenic suppressors with compensatory mutations within the PH domain. These show partial restoration of in vitro preferential PI(4,5)P(2) binding and presence of more motors on pre-synaptic vesicles in vivo. These animals show improved locomotion dependent on in vivo PI(4,5)P(2) levels, increased anterograde transport, and partial restoration of UNC-104 protein levels in vivo. For further proof, we mutated a conserved residue in one suppressor background. The PH domain in this triple mutant lacked in vitro PI(4,5)P(2) binding specificity, and the animals again showed locomotory defects and reduced motor levels. All allelic variants show increased UNC-104 levels upon blocking the ubiquitin pathway. These data show that inability to bind cargo can target motors for degradation. In view of the observed degradation of the motor in synaptic regions, this further suggests that UNC-104 may get degraded at synapses upon release of cargo.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Cinesinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Processamento de Proteína Pós-Traducional , Alelos , Animais , Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans/química , Sequência Conservada/genética , Genes Supressores , Proteínas de Fluorescência Verde/metabolismo , Cinesinas/química , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/química , Neurônios/citologia , Neurônios/metabolismo , Fenótipo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Sinapses/metabolismo , Vesículas Sinápticas/metabolismo , Ubiquitina/metabolismo
9.
Genetics ; 224(3)2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37079426

RESUMO

I outline a streamlined method to insert large, single-copy transgenes into the Caenorhabditis elegans genome using recombination-mediated cassette exchange (RMCE) that relies solely on drug selection yielding a homozygous fluorescent protein (FP) marked transgene in 3 generations (8 days) at high efficiency (>1 insertion per 2 injected P0 animals). Landing sites for this approach are available on four chromosomes in several configurations which yield lines marked in distinct cell types. An array of vectors permit creating transgenes using a variety of selection methods (HygR, NeoR, PuroR, and unc-119) that yield lines expressing different colored FPs (BFP, GFP, mNG, and Scarlet). Although these transgenes retain a plasmid backbone and a selection marker, the inclusion of these sequences typically does not alter the expression of several cell-specific promoters tested. However, in certain orientations, promoters exhibit crosstalk with adjacent transcription units. In cases where crosstalk is problematic, the loxP-flanked fluorescent marker, plasmid backbone, and hygR gene can be excised by crossing through germline Cre expressing lines also created using this technique. Finally, genetic and molecular reagents designed to facilitate customization of both targeting vectors and landing sites are also described. Together, the rapid RMCE toolbox provides a platform for developing further innovative uses of RMCE to create complex genetically engineered tools.


Assuntos
Caenorhabditis elegans , Engenharia Genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Transgenes , Engenharia Genética/métodos , Genoma , Recombinação Genética , Integrases/genética
10.
Sci Adv ; 9(44): eadh2584, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37910615

RESUMO

The γ-aminobutyric acid-mediated (GABAergic) system participates in many aspects of organismal physiology and disease, including proteostasis, neuronal dysfunction, and life-span extension. Many of these phenotypes are also regulated by reactive oxygen species (ROS), but the redox mechanisms linking the GABAergic system to these phenotypes are not well defined. Here, we report that GABAergic redox signaling cell nonautonomously activates many stress response pathways in Caenorhabditis elegans and enhances vulnerability to proteostasis disease in the absence of oxidative stress. Cell nonautonomous redox activation of the mitochondrial unfolded protein response (mitoUPR) proteostasis network requires UNC-49, a GABAA receptor that we show is activated by hydrogen peroxide. MitoUPR induction by a spinocerebellar ataxia type 3 (SCA3) C. elegans neurodegenerative disease model was similarly dependent on UNC-49 in C. elegans. These results demonstrate a multi-tissue paradigm for redox signaling in the GABAergic system that is transduced via a GABAA receptor to function in cell nonautonomous regulation of health, proteostasis, and disease.


Assuntos
Proteínas de Caenorhabditis elegans , Doenças Neurodegenerativas , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Oxirredução , Receptores de GABA-A/metabolismo , Resposta a Proteínas não Dobradas
11.
Neuron ; 55(2): 231-45, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17640525

RESUMO

Upon arriving at their targets, developing axons cease pathfinding and begin instead to arborize and form synapses. To test whether CNS arborization and synaptogenesis are controlled by Slit-Robo signaling, we followed single retinal ganglion cell (RGC) arbors over time. ast (robo2) mutant and slit1a morphant arbors had more branch tips and greater arbor area and complexity compared to wild-type and concomitantly more presumptive presynaptic sites labeled with YFP-Rab3. Increased arborization in ast was phenocopied by dominant-negative Robo2 expressed in single RGCs and rescued by full-length Robo2, indicating that Robo2 acts cell-autonomously. Time-lapse imaging revealed that ast and slit1a morphant arbors stabilized earlier than wild-type, suggesting a role for Slit-Robo signaling in preventing arbor maturation. Genetic analysis showed that Slit1a acts both through Robo2 and Robo2-independent mechanisms. Unlike previous PNS studies showing that Slits promote branching, our results show that Slits inhibit arborization and synaptogenesis in the CNS.


Assuntos
Dendritos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Células Ganglionares da Retina/metabolismo , Sinapses/metabolismo , Vias Visuais/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Células Ganglionares da Retina/citologia , Colículos Superiores/citologia , Colículos Superiores/metabolismo , Vias Visuais/citologia , Peixe-Zebra
12.
J Neurosci ; 30(36): 11951-61, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20826659

RESUMO

Astroglia secrete factors that promote synapse formation and maintenance. In culture, glial contact has also been shown to facilitate synaptogenesis. Here, we examined whether glial contact is important for establishing circuits in vivo by simultaneously monitoring differentiation of glial cells and local synaptogenesis over time. Photoreceptor circuits of the vertebrate retina are particularly suitable for this study because of the relatively simple, laminar organization of their connectivity with their target neurons, horizontal cells and bipolar cells. Also, individual photoreceptor terminals are ensheathed within the outer plexiform layer (OPL) by the processes of one type of glia, Müller glia cells (MGs). We conducted in vivo time-lapse multiphoton imaging of the rapidly developing and relatively transparent zebrafish retina to ascertain the time course of MG development relative to OPL synaptogenesis. The emergence of synaptic triads, indicative of functional photoreceptor circuits, and structural association with glial processes were also examined across ages by electron microscopy. We first show that MG processes form territories that tile within the inner and outer synaptic layers. We then demonstrate that cone photoreceptor synapses are assembled before the elaboration of MG processes in the OPL. Using a targeted cell ablation approach, we also determined whether the maintenance of photoreceptor synapses is perturbed when local MGs are absent. We found that removal of MGs had no appreciable effect on the stability of newly formed cone synapses. Thus, in contrast to other CNS circuits, contact from glia is not necessary for the formation or immediate stabilization of outer retinal synapses.


Assuntos
Neuroglia/fisiologia , Neurônios/fisiologia , Retina/citologia , Sinapses/fisiologia , Aminoácidos , Animais , Animais Geneticamente Modificados , Embrião não Mamífero , Imageamento Tridimensional/métodos , Proteínas Luminescentes/genética , Microscopia Confocal/métodos , Microscopia Eletrônica de Transmissão/métodos , Neuroglia/ultraestrutura , Neurônios/classificação , Neurônios/ultraestrutura , Fotodegradação , Receptores de Glutamato/metabolismo , Sinapses/ultraestrutura , Fatores de Tempo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
13.
Anesthesiology ; 115(6): 1162-71, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22024713

RESUMO

BACKGROUND: Volatile general anesthetics inhibit neurotransmitter release by a mechanism not fully understood. Genetic evidence in Caenorhabditis elegans has shown that a major mechanism of action of volatile anesthetics acting at clinical concentrations in this animal is presynaptic inhibition of neurotransmission. To define additional components of this presynaptic volatile anesthetic mechanism, C. elegans mutants isolated as phenotypic suppressors of a mutation in syntaxin, an essential component of the neurotransmitter release machinery, were screened for anesthetic sensitivity phenotypes. METHODS: Sensitivity to isoflurane concentrations was measured in locomotion assays on adult C. elegans. Sensitivity to the acetylcholinesterase inhibitor aldicarb was used as an assay for the global level of C. elegans acetylcholine release. Comparisons of isoflurane sensitivity (measured by the EC50) were made by simultaneous curve-fitting and F test. RESULTS: Among the syntaxin suppressor mutants, js127 was the most isoflurane resistant, with an EC50 more than 3-fold that of wild type. Genetic mapping, sequencing, and transformation phenocopy showed that js127 was an allele of acy-1, which encodes an adenylate cyclase expressed throughout the C. elegans nervous system and in muscle. js127 behaved as a gain-of-function mutation in acy-1 and had increased concentrations of cyclic adenosine monophosphate. Testing of single and double mutants along with selective tissue expression of the js127 mutation revealed that acy-1 acts in neurons within a Gαs-PKA-UNC-13-dependent pathway to regulate behavior and isoflurane sensitivity. CONCLUSIONS: Activation of neuronal adenylate cyclase antagonizes isoflurane inhibition of locomotion in C. elegans.


Assuntos
Adenilil Ciclases/genética , Caenorhabditis elegans/genética , Resistência a Medicamentos/genética , Isoflurano/farmacologia , Mutação/genética , Proteínas Qa-SNARE/genética , Acetilcolina/metabolismo , Adenilil Ciclases/metabolismo , Aldicarb/metabolismo , Aldicarb/farmacologia , Alelos , Anestésicos Inalatórios/metabolismo , Anestésicos Inalatórios/farmacologia , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/metabolismo , Mapeamento Cromossômico/métodos , Isoflurano/metabolismo , Proteínas Qa-SNARE/efeitos dos fármacos , Proteínas Qa-SNARE/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética
14.
Proc Natl Acad Sci U S A ; 105(42): 16350-5, 2008 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-18852466

RESUMO

The Caenorhabditis elegans defecation motor program (DMP) is a highly coordinated rhythmic behavior that requires two GABAergic neurons that synapse onto the enteric muscles. One class of DMP mutants, called anterior body wall muscle contraction and expulsion defective (aex) mutants, exhibits similar defects to those caused by the loss of these two neurons. Here, we demonstrate that aex-2 encodes a G-protein-coupled receptor (GPCR) and aex-4 encodes an exocytic SNAP25 homologue. We found that aex-2 functions in the nervous system and activates a G(s)alpha signaling pathway to regulate defecation. aex-4, on the other hand, functions in the intestinal epithelial cells. Furthermore, we show that aex-5, which encodes a pro-protein convertase, functions in the intestine to regulate the DMP and that its secretion from the intestine is impaired in aex-4 mutants. Activation of the G(s)alpha GPCR pathway in GABAergic neurons can suppress the defecation defect of the intestinal mutants aex-4 and aex-5. Lastly, we demonstrate that activation of GABAergic neurons using the light-gated cation channel channelrhodopsin-2 is sufficient to suppress the behavioral defects of aex-2, aex-4, and aex-5. These results genetically place intestinal genes aex-4 and aex-5 upstream of GABAergic GPCR signaling. We propose a model whereby the intestinal genes aex-4 and aex-5 control the DMP by regulating the secretion of a signal, which activates the neuronal receptor aex-2.


Assuntos
Comportamento Animal/fisiologia , Caenorhabditis elegans/citologia , Caenorhabditis elegans/fisiologia , Mucosa Intestinal/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo , Animais , Regulação da Expressão Gênica , Luz , Locomoção , Dados de Sequência Molecular , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas SNARE/metabolismo , Ácido gama-Aminobutírico/biossíntese
15.
Nat Neurosci ; 9(12): 1479-87, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17115037

RESUMO

A central event in synapse development is formation of the presynaptic active zone in response to positional cues. Three active zone proteins, RIM, ELKS (also known as ERC or CAST) and Liprin-alpha, bind each other and are implicated in linking active zone formation to synaptic vesicle release. Loss of function in Caenorhabditis elegans syd-2 Liprin-alpha alters the size of presynaptic specializations and disrupts synaptic vesicle accumulation. Here we report that a missense mutation in the coiled-coil domain of SYD-2 causes a gain of function. In HSN synapses, the syd-2(gf) mutation promotes synapse formation in the absence of syd-1, which is essential for HSN synapse formation. syd-2(gf) also partially suppresses the synaptogenesis defects in syg-1 and syg-2 mutants. The activity of syd-2(gf) requires elks-1, an ELKS homolog; but not unc-10, a RIM homolog. The mutant SYD-2 shows increased association with ELKS. These results establish a functional dependency for assembly of the presynaptic active zone in which SYD-2 plays a key role.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Transporte/metabolismo , Fosfoproteínas/fisiologia , Terminações Pré-Sinápticas/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiologia , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Neurônios Motores/metabolismo , Mutação , Fosfoproteínas/genética , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
16.
Genetics ; 215(4): 903-921, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32513816

RESUMO

The application of CRISPR technology has greatly facilitated the creation of transgenic Caenorhabditis elegans lines. However, methods to insert multi-kilobase DNA constructs remain laborious even with these advances. Here, I describe a new approach for introducing large DNA constructs into the C. elegans genome at specific sites using a combination of Flp and Cre recombinases. The system utilizes specialized integrated landing sites that express GFP ubiquitously flanked by single loxP, FRT, and FRT3 sites. DNA sequences of interest are inserted into an integration vector that contains a sqt-1 self-excising cassette and FRT and FRT3 sites. Plasmid DNA is injected into the germline of landing site animals. Transgenic animals are identified as Rol progeny, and the sqt-1 marker is subsequently excised with heat shock Cre expression. Integration events were obtained at a rate of approximately one integration per three injected F0 animals-a rate substantially higher than any current approach. To demonstrate the robustness of the approach, I compared the efficiency of the Gal4/UAS, QF (and QF2)/QUAS, tetR(and rtetR)/tetO, and LexA/lexO bipartite expression systems by assessing expression levels in combinations of driver and reporter GFP constructs and a direct promoter GFP fusion each integrated at multiple sites in the genome. My data demonstrate that all four bipartite systems are functional in C. elegans Although the new integration system has several limitations, it greatly reduces the effort required to create single-copy insertions at defined sites in the C. elegans genome.


Assuntos
Animais Geneticamente Modificados/fisiologia , Caenorhabditis elegans/fisiologia , DNA Nucleotidiltransferases/metabolismo , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/metabolismo , Plasmídeos/genética , Recombinação Genética , Animais , Animais Geneticamente Modificados/genética , Caenorhabditis elegans/genética , DNA Nucleotidiltransferases/genética , Proteínas de Fluorescência Verde/genética , Regiões Promotoras Genéticas
17.
Biochemistry ; 48(22): 4663-5, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19397310

RESUMO

Despite much work, subcellular neurons of Caenorhabditis elegans have not been studied at nanometer resolution with millisecond time resolution. Nor has there been an effective way to immobilize C. elegans. Here we show that, without using anesthetic or paralyzing agents, fluorescence imaging with one-nanometer accuracy (FIONA) can be successfully applied to fluorescently labeled molecules within C. elegans nerves. GFP- and DENDRA2-labeled ELKS punctae can be localized with sub-10 nm accuracy in approximately 5 ms. Our results show that the protein ELKS is occasionally transferred by microtubule-based motors. This is the first example of FIONA applied to a living organism.


Assuntos
Proteínas de Caenorhabditis elegans/isolamento & purificação , Proteínas de Transporte/isolamento & purificação , Proteínas do Tecido Nervoso/isolamento & purificação , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Células Cultivadas , Corantes Fluorescentes/química , Proteínas de Fluorescência Verde/química , Peptídeos e Proteínas de Sinalização Intracelular , Quimografia , Proteínas Motores Moleculares/isolamento & purificação , Proteínas Motores Moleculares/metabolismo , Proteínas Motores Moleculares/fisiologia , Neurônios Motores/química , Neurônios Motores/fisiologia , Movimento/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia
18.
PLoS Biol ; 4(8): e261, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16895441

RESUMO

Caenorhabditis elegans TOM-1 is orthologous to vertebrate tomosyn, a cytosolic syntaxin-binding protein implicated in the modulation of both constitutive and regulated exocytosis. To investigate how TOM-1 regulates exocytosis of synaptic vesicles in vivo, we analyzed C. elegans tom-1 mutants. Our electrophysiological analysis indicates that evoked postsynaptic responses at tom-1 mutant synapses are prolonged leading to a two-fold increase in total charge transfer. The enhanced response in tom-1 mutants is not associated with any detectable changes in postsynaptic response kinetics, neuronal outgrowth, or synaptogenesis. However, at the ultrastructural level, we observe a concomitant increase in the number of plasma membrane-contacting vesicles in tom-1 mutant synapses, a phenotype reversed by neuronal expression of TOM-1. Priming defective unc-13 mutants show a dramatic reduction in plasma membrane-contacting vesicles, suggesting these vesicles largely represent the primed vesicle pool at the C. elegans neuromuscular junction. Consistent with this conclusion, hyperosmotic responses in tom-1 mutants are enhanced, indicating the primed vesicle pool is enhanced. Furthermore, the synaptic defects of unc-13 mutants are partially suppressed in tom-1 unc-13 double mutants. These data indicate that in the intact nervous system, TOM-1 negatively regulates synaptic vesicle priming.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Vesículas Sinápticas/fisiologia , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/análise , Proteínas de Caenorhabditis elegans/genética , Eletrofisiologia , Dados de Sequência Molecular , Mutação , Junção Neuromuscular/fisiologia , Fenótipo , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Alinhamento de Sequência , Vesículas Sinápticas/química , Vesículas Sinápticas/ultraestrutura
19.
Mol Biol Cell ; 17(6): 2617-25, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16571673

RESUMO

Rab small GTPases are involved in the transport of vesicles between different membranous organelles. RAB-3 is an exocytic Rab that plays a modulatory role in synaptic transmission. Unexpectedly, mutations in the Caenorhabditis elegans RAB-3 exchange factor homologue, aex-3, cause a more severe synaptic transmission defect as well as a defecation defect not seen in rab-3 mutants. We hypothesized that AEX-3 may regulate a second Rab that regulates these processes with RAB-3. We found that AEX-3 regulates another exocytic Rab, RAB-27. Here, we show that C. elegans RAB-27 is localized to synapse-rich regions pan-neuronally and is also expressed in intestinal cells. We identify aex-6 alleles as containing mutations in rab-27. Interestingly, aex-6 mutants exhibit the same defecation defect as aex-3 mutants. aex-6; rab-3 double mutants have behavioral and pharmacological defects similar to aex-3 mutants. In addition, we demonstrate that RBF-1 (rabphilin) is an effector of RAB-27. Therefore, our work demonstrates that AEX-3 regulates both RAB-3 and RAB-27, that both RAB-3 and RAB-27 regulate synaptic transmission, and that RAB-27 potentially acts through its effector RBF-1 to promote soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) function.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Transmissão Sináptica/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo , Animais , Animais Geneticamente Modificados , Sequência de Bases , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Clonagem Molecular , Primers do DNA , Proteínas de Fluorescência Verde/genética , Guanosina Trifosfato/metabolismo , Mutação , Regiões Promotoras Genéticas , Proteínas SNARE/metabolismo
20.
Neuron ; 33(2): 155-6, 2002 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-11804562

RESUMO

A variety of secreted components have been identified as retrograde signals mediating diverse aspects of synaptic development, maintenance, and plasticity; however, little is known about the mechanisms mediating the release of secreted retrograde signals. Doi and Iwasaki (this issue of Neuron) implicate AEX-1, a protein distantly related to the UNC-13/Munc13 family, as an attractive candidate regulator of the retrograde release machinery in muscle.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas do Tecido Nervoso/fisiologia , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Animais , Caenorhabditis elegans/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa