Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38963411

RESUMO

Precise developmental timing control is essential for organism formation and function, but its mechanisms are unclear. In C. elegans, the microRNA lin-4 critically regulates developmental timing by post-transcriptionally downregulating the larval-stage-fate controller LIN-14. However, the mechanisms triggering the activation of lin-4 expression toward the end of the first larval stage remain unknown. We demonstrate that the transmembrane transcription factor MYRF-1 is necessary for lin-4 activation. MYRF-1 is initially localized on the cell membrane, and its increased cleavage and nuclear accumulation coincide with lin-4 expression timing. MYRF-1 regulates lin-4 expression cell-autonomously and hyperactive MYRF-1 can prematurely drive lin-4 expression in embryos and young first-stage larvae. The tandem lin-4 promoter DNA recruits MYRF-1GFP to form visible loci in the nucleus, suggesting that MYRF-1 directly binds to the lin-4 promoter. Our findings identify a crucial link in understanding developmental timing regulation and establish MYRF-1 as a key regulator of lin-4 expression.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs , Fatores de Transcrição , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crescimento & desenvolvimento , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , MicroRNAs/metabolismo , MicroRNAs/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Regiões Promotoras Genéticas , Transcrição Gênica , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Núcleo Celular/metabolismo
2.
Bioessays ; 46(4): e2300209, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38488284

RESUMO

The Myelin Regulator Factor (MYRF) is a master regulator governing myelin formation and maintenance in the central nervous system. The conservation of MYRF across metazoans and its broad tissue expression suggest it has functions extending beyond the well-established role in myelination. Loss of MYRF results in developmental lethality in both invertebrates and vertebrates, and MYRF haploinsufficiency in humans causes MYRF-related Cardiac Urogenital Syndrome, underscoring its importance in animal development; however, these mechanisms are largely unexplored. MYRF, an unconventional transcription factor, begins embedded in the membrane and undergoes intramolecular chaperone mediated trimerization, which triggers self-cleavage, allowing its N-terminal segment with an Ig-fold DNA-binding domain to enter the nucleus for transcriptional regulation. Recent research suggests developmental regulation of cleavage, yet the mechanisms remain enigmatic. While some parts of MYRF's structure have been elucidated, others remain obscure, leaving questions about how these motifs are linked to its intricate processing and function.


Assuntos
Bainha de Mielina , Fatores de Transcrição , Animais , Humanos , Fatores de Transcrição/metabolismo , Bainha de Mielina/metabolismo , Proteínas de Membrana/metabolismo , Regulação da Expressão Gênica , Domínios Proteicos
3.
Cell Rep ; 42(10): 113246, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37831605

RESUMO

Metastasis is the leading cause of high ovarian-cancer-related mortality worldwide. Three major processes constitute the whole metastatic cascade: invasion, intravasation, and extravasation. Tumor cells often reprogram their metabolism to gain advantages in proliferation and survival. However, whether and how those metabolic alterations contribute to the invasiveness of tumor cells has yet to be fully understood. Here we performed a genome-wide CRISPR-Cas9 screening to identify genes participating in tumor cell dissemination and revealed that PTGES3 acts as an invasion suppressor in ovarian cancer. Mechanistically, PTGES3 binds to phosphofructokinase, liver type (PFKL) and generates a local source of prostaglandin E2 (PGE2) to allosterically inhibit the enzymatic activity of PFKL. Repressed PFKL leads to downgraded glycolysis and the subsequent TCA cycle for glucose metabolism. However, ovarian cancer suppresses the expression of PTGES3 and disrupts the PTGES3-PGE2-PFKL inhibitory axis, leading to hyperactivation of glucose oxidation, eventually facilitating ovarian cancer cell motility and invasiveness.


Assuntos
Dinoprostona , Neoplasias Ovarianas , Humanos , Feminino , Fosfofrutoquinases , Fosfofrutoquinase-1/genética , Fígado/metabolismo , Glucose/metabolismo , Neoplasias Ovarianas/patologia , Proliferação de Células , Linhagem Celular Tumoral , Invasividade Neoplásica
4.
Elife ; 102021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33950834

RESUMO

Neural circuits develop through a plastic phase orchestrated by genetic programs and environmental signals. We have identified a leucine-rich-repeat domain transmembrane protein PAN-1 as a factor required for synaptic rewiring in C. elegans. PAN-1 localizes on cell membrane and binds with MYRF, a membrane-bound transcription factor indispensable for promoting synaptic rewiring. Full-length MYRF was known to undergo self-cleavage on ER membrane and release its transcriptional N-terminal fragment in cultured cells. We surprisingly find that MYRF trafficking to cell membrane before cleavage is pivotal for C. elegans development and the timing of N-MYRF release coincides with the onset of synaptic rewiring. On cell membrane PAN-1 and MYRF interact with each other via their extracellular regions. Loss of PAN-1 abolishes MYRF cell membrane localization, consequently blocking myrf-dependent neuronal rewiring process. Thus, through interactions with a cooperating factor on the cell membrane, MYRF may link cell surface activities to transcriptional cascades required for development.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/química , Fatores de Transcrição/metabolismo , Animais , Sistemas CRISPR-Cas , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Membrana Celular/metabolismo , Células HEK293 , Humanos , Domínios Proteicos , Transporte Proteico , Sinapses/fisiologia , Fatores de Transcrição/genética
5.
iScience ; 11: 224-237, 2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30634168

RESUMO

Binary gene regulatory tools such as the Tetracycline (Tet)-controlled transcription system have revolutionized genetic research in multiple organisms, but their applications to the worm remain very limited. Here we report that the canonical Tet system is largely inactive in the worm but can be adapted for the worm by introducing multiple modifications, a crucial one being the use of the transcription activation domain from the fungal Q binary system. The resultant Tet/Q hybrid system proves more robust and flexible than either of its precursors, enabling elaborate modes of transgene manipulation previously hard to achieve in the worm, including inducible intersectional regulation and, in combination with the Q system, independent control of distinct transgenes within the same cells. Furthermore, we demonstrated, as an example of its applications, that the hybrid system can tightly and efficiently control Cre expression. This study establishes Tet/Q as a premier binary system for worm genetic research.

6.
J Neurosci ; 38(46): 9829-9839, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30249802

RESUMO

Myelin sheath formed by oligodendrocytes (OLs) is essential for the rapid propagation of action potentials in the vertebrate CNS. Myelin regulatory factor (MYRF) is one of the critical factors that control OL differentiation and myelin maintenance. Previous studies showed that MYRF is a membrane-bound transcription factor associated with the endoplasmic reticulum (ER). After self-cleavage, the N-fragment of MYRF is released from the ER and translocated into the nucleus where it functions as a transcription factor to activate myelin gene expression. At present, it remains unknown whether MYRF self-cleavage and functional activation can be regulated during OL differentiation. Here, we report that TMEM98, an ER-associated transmembrane protein, is capable of binding to the C-terminal of MYRF and inhibiting its self-cleavage and N-fragment nuclear translocation. In the developing CNS, TMEM98 is selectively expressed in early maturing OLs in mouse pups of either sex. Forced expression of TMEM98 in embryonic chicken spinal cord of either sex suppresses endogenous OL differentiation and MYRF-induced ectopic expression of myelin genes. These results suggest that TMEM98, through inhibiting the self-cleavage of MYRF, functions as a negative feedback regulator of MYRF in oligodendrocyte differentiation and myelination.SIGNIFICANCE STATEMENT MYRF protein is initially synthesized as an ER-associated membrane protein that undergoes autoproteolytic cleavage to release the N-fragment, which is then transported into the nucleus and activates the transcription of myelin genes. To date, the molecular mechanisms that regulate the self-cleavage and function of MYRF in regulating oligodendrocyte differentiation have remained unknown. In this study, we present the molecular and functional evidence that TMEM98 membrane protein physically interacts with MYRF in the ER and subsequently blocks its self-cleavage, N-terminal nuclear translocation, and functional activation of myelin gene expression. To our knowledge, this is the first report on the regulation of MYRF self-proteolytic activity and function by an interacting protein, providing new insights into the molecular regulation of OL differentiation and myelinogenesis.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Membrana/metabolismo , Oligodendroglia/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Galinhas , Retículo Endoplasmático/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Ligação Proteica/fisiologia
7.
Elife ; 72018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29360035

RESUMO

Cell- or network-driven oscillators underlie motor rhythmicity. The identity of C. elegans oscillators remains unknown. Through cell ablation, electrophysiology, and calcium imaging, we show: (1) forward and backward locomotion is driven by different oscillators; (2) the cholinergic and excitatory A-class motor neurons exhibit intrinsic and oscillatory activity that is sufficient to drive backward locomotion in the absence of premotor interneurons; (3) the UNC-2 P/Q/N high-voltage-activated calcium current underlies A motor neuron's oscillation; (4) descending premotor interneurons AVA, via an evolutionarily conserved, mixed gap junction and chemical synapse configuration, exert state-dependent inhibition and potentiation of A motor neuron's intrinsic activity to regulate backward locomotion. Thus, motor neurons themselves derive rhythms, which are dually regulated by the descending interneurons to control the reversal motor state. These and previous findings exemplify compression: essential circuit properties are conserved but executed by fewer numbers and layers of neurons in a small locomotor network.


Assuntos
Relógios Biológicos , Caenorhabditis elegans/fisiologia , Locomoção , Neurônios Motores/fisiologia , Periodicidade , Animais , Neurônios Colinérgicos/fisiologia , Interneurônios/fisiologia
8.
Curr Opin Neurobiol ; 48: 97-105, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29182952

RESUMO

The ability of neurons to modify or remodel their synaptic connectivity is critical for the function of neural circuitry throughout the life of an animal. Understanding the mechanisms underlying neuronal structural changes is central to our knowledge of how the nervous system is shaped for complex behaviors and how it further adapts to developmental and environmental demands. Caenorhabditis elegans provides a powerful model for examining developmental processes and for discovering mechanisms controlling neural plasticity. Recent findings have identified conserved themes underlying neural plasticity in development and under environmental stress.


Assuntos
Comportamento Animal/fisiologia , Sistema Nervoso/citologia , Sistema Nervoso/crescimento & desenvolvimento , Neurônios/fisiologia , Sinapses/fisiologia , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Mutação/genética , Neuroglia/fisiologia
9.
G3 (Bethesda) ; 7(7): 2055-2063, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28468816

RESUMO

Acetylcholine (ACh) receptors (AChR) regulate neural circuit activity in multiple contexts. In humans, mutations in ionotropic acetylcholine receptor (iAChR) genes can cause neurological disorders, including myasthenia gravis and epilepsy. In Caenorhabditis elegans, iAChRs play multiple roles in the locomotor circuit. The cholinergic motor neurons express an ACR-2-containing pentameric AChR (ACR-2R) comprised of ACR-2, ACR-3, ACR-12, UNC-38, and UNC-63 subunits. A gain-of-function mutation in the non-α subunit gene acr-2 [acr-2(gf)] causes defective locomotion as well as spontaneous convulsions. Previous studies of genetic suppressors of acr-2(gf) have provided insights into ACR-2R composition and assembly. Here, to further understand how the ACR-2R regulates neuronal activity, we expanded the suppressor screen for acr-2(gf)-induced convulsions. The majority of these suppressor mutations affect genes that play critical roles in synaptic transmission, including two novel mutations in the vesicular ACh transporter unc-17 In addition, we identified a role for a conserved major facilitator superfamily domain (MFSD) protein, mfsd-6, in regulating neural circuit activity. We further defined a role for the sphingosine (SPH) kinase (Sphk) sphk-1 in cholinergic neuron activity, independent of previously known signaling pathways. Overall, the genes identified in our study suggest that optimal modulation of synaptic activity is balanced by the differential activities of multiple pathways, and the novel alleles provide valuable reagents to further dissect neuronal mechanisms regulating the locomotor circuit.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Locomoção/genética , Mutação , Receptores Colinérgicos , Transmissão Sináptica/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Neurônios Colinérgicos/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo
10.
Dev Cell ; 41(2): 180-194.e7, 2017 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-28441531

RESUMO

Synaptic refinement is a critical step in nervous system maturation, requiring a carefully timed reorganization and refinement of neuronal connections. We have identified myrf-1 and myrf-2, two C. elegans homologs of Myrf family transcription factors, as key regulators of synaptic rewiring. MYRF-1 and its paralog MYRF-2 are functionally redundant specifically in synaptic rewiring. They co-exist in the same protein complex and act cooperatively to regulate synaptic rewiring. We find that the MYRF proteins localize to the ER membrane and that they are cleaved into active N-terminal fragments, which then translocate into the nucleus to drive synaptic rewiring. Overexpression of active forms of MYRF is sufficient to accelerate synaptic rewiring. MYRF-1 and MYRF-2 are the first genes identified to be indispensable for promoting synaptic rewiring in C. elegans. These findings reveal a molecular mechanism underlying synaptic rewiring and developmental circuit plasticity.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Retículo Endoplasmático/metabolismo , Plasticidade Neuronal/genética , Sinapses/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/economia , Proteínas de Caenorhabditis elegans/metabolismo , Núcleo Celular/metabolismo , Proteínas de Membrana/metabolismo
11.
Genes Dev ; 29(22): 2377-90, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26588990

RESUMO

Alternative polyadenylation (APA) is widespread in neuronal development and activity-mediated neural plasticity. However, the underlying molecular mechanisms are largely unknown. We used systematic genetic studies and genome-wide surveys of the transcriptional landscape to identify a context-dependent regulatory pathway controlling APA in the Caenorhabditis elegans nervous system. Loss of function in ssup-72, a Ser5 phosphatase for the RNA polymerase II (Pol II) C-terminal domain (CTD), dampens transcription termination at a strong intronic polyadenylation site (PAS) in unc-44/ankyrin yet promotes termination at the weak intronic PAS of the MAP kinase dlk-1. A nuclear protein, SYDN-1, which regulates neuronal development, antagonizes the function of SSUP-72 and several nuclear polyadenylation factors. This regulatory pathway allows the production of a neuron-specific isoform of unc-44 and an inhibitory isoform of dlk-1. Dysregulation of the unc-44 and dlk-1 mRNA isoforms in sydn-1 mutants impairs neuronal development. Deleting the intronic PAS of unc-44 results in increased pre-mRNA processing of neuronal ankyrin and suppresses sydn-1 mutants. These results reveal a mechanism by which regulation of CTD phosphorylation controls coding region APA in the nervous system.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/crescimento & desenvolvimento , Fosfoproteínas Fosfatases/metabolismo , Animais , Anquirinas/genética , Anquirinas/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Regulação da Expressão Gênica no Desenvolvimento , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Mutação , Neurônios/enzimologia , Fosfoproteínas Fosfatases/genética , Poliadenilação , Ligação Proteica
12.
Biotechnol Biotechnol Equip ; 29(1): 105-110, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-26740790

RESUMO

Traditional cut-paste DNA cloning is often limited by the availability of restriction enzyme sites. Here, we described the complementary annealing mediated by exonuclease (CAME), in which the insert or vector fragment is amplified to carry sequences complementary to the other, and both fragments are modified by exonuleases to create directional single-stranded overhangs. The two recessed DNA fragments are joined through complementary strand annealing. The CAME is highly efficient for cloning the DNA of at least 12 kb and single DNA fragment out of a complex DNA sample. Moreover, the application of CAME greatly improved the efficiency of site-directed mutagenesis.

13.
J Neurosci ; 33(12): 5319-25, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23516296

RESUMO

Excitatory acetylcholine motor neurons drive Caenorhabditis elegans locomotion. Coordinating the activation states of the backward-driving A and forward-driving B class motor neurons is critical for generating sinusoidal and directional locomotion. Here, we show by in vivo calcium imaging that expression of a hyperactive, somatodendritic ionotropic acetylcholine receptor ACR-2(gf) in A and B class motor neurons induces aberrant synchronous activity in both ventral- and dorsal-innervating B and A class motor neurons. Expression of ACR-2(gf) in either ventral- or dorsal-innervating B neurons is sufficient for triggering the aberrant synchrony that results in arrhythmic convulsions. Silencing of AVB, the premotor interneurons that innervate B motor neurons suppresses ACR-2(gf)-dependent convulsion; activating AVB by channelrhodopsin induces the onset of convulsion. These results support that the activity state of B motor neurons plays an instructive role for the coordination of motor circuit.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Neurônios Colinérgicos/fisiologia , Vias Eferentes/citologia , Vias Eferentes/fisiologia , Neurônios Motores/fisiologia , Receptores Nicotínicos/fisiologia , Acetilcolina/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Dendritos/fisiologia , Expressão Gênica/fisiologia , Interneurônios/fisiologia , Locomoção/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores Nicotínicos/genética , Convulsões/genética , Convulsões/fisiopatologia
14.
Bio Protoc ; 3(1)2013 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27525293

RESUMO

This protocol describes a method for light-inducible cell ablation in live worms. miniSOG (mini Singlet Oxygen Generator) generates singlet oxygen upon blue light illumination (Shu et al., 2011). Mitochondrially membrane targeted miniSOG (the first 55 a. a. of C. e. tomm-20 fused at the N'-terminus of miniSOG, termed as mito-miniSOG in the following) is transgenically expressed in specific cells/tissues (Qi et al., 2012). Groups of transgenic animals are illuminated under open field fluorescence light on a compound microscope or LED light setup for photo-ablation.

15.
Proc Natl Acad Sci U S A ; 109(19): 7499-504, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22532663

RESUMO

We describe a method for light-inducible and tissue-selective cell ablation using a genetically encoded photosensitizer, miniSOG (mini singlet oxygen generator). miniSOG is a newly engineered fluorescent protein of 106 amino acids that generates singlet oxygen in quantum yield upon blue-light illumination. We transgenically expressed mitochondrially targeted miniSOG (mito-miniSOG) in Caenorhabditis elegans neurons. Upon blue-light illumination, mito-miniSOG causes rapid and effective death of neurons in a cell-autonomous manner without detectable damages to surrounding tissues. Neuronal death induced by mito-miniSOG appears to be independent of the caspase CED-3, but the clearance of the damaged cells partially depends on the phagocytic receptor CED-1, a homolog of human CD91. We show that neurons can be killed at different developmental stages. We further use this method to investigate the role of the premotor interneurons in regulating the convulsive behavior caused by a gain-of-function mutation in the neuronal acetylcholine receptor acr-2. Our findings support an instructive role for the interneuron AVB in controlling motor neuron activity and reveal an inhibitory effect of the backward premotor interneurons on the forward interneurons. In summary, the simple inducible cell ablation method reported here allows temporal and spatial control and will prove to be a useful tool in studying the function of specific cells within complex cellular contexts.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Flavoproteínas/metabolismo , Proteínas Luminescentes/metabolismo , Neurônios/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/citologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Caspases/genética , Caspases/metabolismo , Morte Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Flavoproteínas/genética , Interneurônios/citologia , Interneurônios/metabolismo , Interneurônios/efeitos da radiação , Luz , Proteínas Luminescentes/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neurônios Motores/efeitos da radiação , Neurônios/citologia , Neurônios/efeitos da radiação , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Oxigênio Singlete/metabolismo , Fatores de Tempo
16.
PLoS Biol ; 9(4): e1001041, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21483721

RESUMO

Electron microscopy (EM) achieves the highest spatial resolution in protein localization, but specific protein EM labeling has lacked generally applicable genetically encoded tags for in situ visualization in cells and tissues. Here we introduce "miniSOG" (for mini Singlet Oxygen Generator), a fluorescent flavoprotein engineered from Arabidopsis phototropin 2. MiniSOG contains 106 amino acids, less than half the size of Green Fluorescent Protein. Illumination of miniSOG generates sufficient singlet oxygen to locally catalyze the polymerization of diaminobenzidine into an osmiophilic reaction product resolvable by EM. MiniSOG fusions to many well-characterized proteins localize correctly in mammalian cells, intact nematodes, and rodents, enabling correlated fluorescence and EM from large volumes of tissue after strong aldehyde fixation, without the need for exogenous ligands, probes, or destructive permeabilizing detergents. MiniSOG permits high quality ultrastructural preservation and 3-dimensional protein localization via electron tomography or serial section block face scanning electron microscopy. EM shows that miniSOG-tagged SynCAM1 is presynaptic in cultured cortical neurons, whereas miniSOG-tagged SynCAM2 is postsynaptic in culture and in intact mice. Thus SynCAM1 and SynCAM2 could be heterophilic partners. MiniSOG may do for EM what Green Fluorescent Protein did for fluorescence microscopy.


Assuntos
Coloração e Rotulagem , 3,3'-Diaminobenzidina/análise , Estruturas Animais , Animais , Encéfalo/ultraestrutura , Caenorhabditis elegans/química , Células , Corantes Fluorescentes/análise , Camundongos , Microscopia Eletrônica , Microscopia de Fluorescência , Mitocôndrias/ultraestrutura , Fototropinas/análise , Proteínas/análise
17.
Development ; 137(13): 2237-50, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20530551

RESUMO

Nuclear pre-mRNA 3'-end processing is vital for the production of mature mRNA and the generation of the 3' untranslated region (UTR). However, the roles and regulation of this event in cellular development remain poorly understood. Here, we report the function of a nuclear pre-mRNA 3'-end processing pathway in synapse and axon formation in C. elegans. In a genetic enhancer screen for synaptogenesis mutants, we identified a novel polyproline-rich protein, Synaptic defective enhancer-1 (SYDN-1). Loss of function of sydn-1 causes abnormal synapse and axon development, and displays striking synergistic interactions with several genes that regulate specific aspects of synapses. SYDN-1 is required in neurons and localizes to distinct regions of the nucleus. Through a genetic suppressor screen, we found that the neuronal defects of sydn-1 mutants are suppressed by loss of function in Polyadenylation factor subunit-2 (PFS-2), a conserved WD40-repeat protein that interacts with multiple subcomplexes of the pre-mRNA 3'-end processing machinery. PFS-2 partially colocalizes with SYDN-1, and SYDN-1 influences the nuclear abundance of PFS-2. Inactivation of several members of the nuclear 3'-end processing complex suppresses sydn-1 mutants. Furthermore, lack of sydn-1 can increase the activity of 3'-end processing. Our studies provide in vivo evidence for pre-mRNA 3'-end processing in synapse and axon development and identify SYDN-1 as a negative regulator of this cellular event in neurons.


Assuntos
Regiões 3' não Traduzidas , Axônios/metabolismo , Caenorhabditis elegans/metabolismo , Sinapses , Animais , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
18.
Glia ; 58(4): 458-68, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19780200

RESUMO

Recent studies have suggested that Nkx6.2/Gtx and Nkx2.2 homeodomain transcription factors are involved in the regulation of oligodendrocyte maturation and/or myelination which occur predominantly in postnatal stages. However, their cellular specificity in postnatal central nervous system has not been characterized and their dynamic expressional relationship during oligodendrocyte lineage progression has not been determined. Here we report that both Nkx2.2 and Nkx6.2 are selectively expressed in Olig2+ cells of oligodendrocyte lineage in postnatal spinal cords. Although Nkx6.2 is specifically expressed in the APC+ mature oligodendrocytes, Nkx2.2 is initially expressed in differentiating oligodendrocyte precursor cells (OPCs) but quickly down-regulated as OPCs undergo terminal differentiation. Intriguingly, Nkx2.2 expression is up-regulated in mature myelinating oligodendrocytes at later stages. The co-expression of Nkx2.2 and Nkx6.2 transcription factors in myelinating oligodendrocytes suggests their functional interactions in the regulation of myelin sheath formation and/or maintenance.


Assuntos
Encéfalo/fisiologia , Proteínas de Homeodomínio/metabolismo , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Medula Espinal/fisiologia , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/citologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/crescimento & desenvolvimento , Células-Tronco/fisiologia , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra
19.
PLoS Biol ; 7(12): e1000265, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20027209

RESUMO

In the nematode Caenorhabditis elegans, cholinergic motor neurons stimulate muscle contraction as well as activate GABAergic motor neurons that inhibit contraction of the contralateral muscles. Here, we describe the composition of an ionotropic acetylcholine receptor that is required to maintain excitation of the cholinergic motor neurons. We identified a gain-of-function mutation that leads to spontaneous muscle convulsions. The mutation is in the pore domain of the ACR-2 acetylcholine receptor subunit and is identical to a hyperactivating mutation in the muscle receptor of patients with myasthenia gravis. Screens for suppressors of the convulsion phenotype led to the identification of other receptor subunits. Cell-specific rescue experiments indicate that these subunits function in the cholinergic motor neurons. Expression of these subunits in Xenopus oocytes demonstrates that the functional receptor is comprised of three alpha-subunits, UNC-38, UNC-63 and ACR-12, and two non-alpha-subunits, ACR-2 and ACR-3. Although this receptor exhibits a partially overlapping subunit composition with the C. elegans muscle acetylcholine receptor, it shows distinct pharmacology. Recordings from intact animals demonstrate that loss-of-function mutations in acr-2 reduce the excitability of the cholinergic motor neurons. By contrast, the acr-2(gf) mutation leads to a hyperactivation of cholinergic motor neurons and an inactivation of downstream GABAergic motor neurons in a calcium dependent manner. Presumably, this imbalance between excitatory and inhibitory input into muscles leads to convulsions. These data indicate that the ACR-2 receptor is important for the coordinated excitation and inhibition of body muscles underlying sinusoidal movement.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Locomoção , Neurônios Motores/metabolismo , Contração Muscular , Receptores Nicotínicos/metabolismo , Acetilcolina/metabolismo , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Dados de Sequência Molecular , Mutação , Receptores Nicotínicos/genética , Transmissão Sináptica , Xenopus , Ácido gama-Aminobutírico/metabolismo
20.
Proc Natl Acad Sci U S A ; 105(7): 2481-6, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18272476

RESUMO

Enhancers have been functionally described for >35 years, but the molecular principles underlying the integration of regulatory inputs to alternate gene enhancers used during mammalian organogenesis remain incompletely understood. Using a combination of in vivo enhancer mapping and proteomics approaches, we have established that two distant and distinct early enhancers, each requiring different transcription complexes, are required for full activation of the gene encoding the pituitary lineage determining factor, Pit1. A transcription factor belonging to the "giant, multiple-homeodomain and zinc finger family," Atbf1, serves as a novel pituitary regulator for one of the two required enhancers as shown by genetic and in vitro analysis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição Pit-1/metabolismo , Animais , Sequência de Bases , Linhagem da Célula , Células-Tronco Embrionárias/metabolismo , Epistasia Genética , Genoma/genética , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutação/genética , Hipófise/metabolismo , Ligação Proteica , Proteômica , Fatores de Tempo , Fator de Transcrição Pit-1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...