Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 7(6): e38569, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701670

RESUMO

Protein ser/thr phosphatase 2A family members (PP2A, PP4, and PP6) are implicated in the control of numerous biological processes, but our understanding of the in vivo function and regulation of these enzymes is limited. In this study, we investigated the role of Tap42, a common regulatory subunit for all three PP2A family members, in the development of Drosophila melanogaster wing imaginal discs. RNAi-mediated silencing of Tap42 using the binary Gal4/UAS system and two disc drivers, pnr- and ap-Gal4, not only decreased survival rates but also hampered the development of wing discs, resulting in a remarkable thorax cleft and defective wings in adults. Silencing of Tap42 also altered multiple signaling pathways (HH, JNK and DPP) and triggered apoptosis in wing imaginal discs. The Tap42(RNAi)-induced defects were the direct result of loss of regulation of Drosophila PP2A family members (MTS, PP4, and PPV), as enforced expression of wild type Tap42, but not a phosphatase binding defective Tap42 mutant, rescued fly survivorship and defects. The experimental platform described herein identifies crucial roles for Tap42•phosphatase complexes in governing imaginal disc and fly development.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Discos Imaginais/enzimologia , Morfogênese/fisiologia , Fosfoproteínas Fosfatases/metabolismo , Transdução de Sinais/fisiologia , Asas de Animais/crescimento & desenvolvimento , Animais , Apoptose/genética , Apoptose/fisiologia , Proteínas de Drosophila/genética , Discos Imaginais/crescimento & desenvolvimento , Imuno-Histoquímica , Interferência de RNA , Transdução de Sinais/genética , Fatores de Transcrição/genética
2.
J Neurogenet ; 26(2): 144-50, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22420370

RESUMO

This is the first of two reviews that include some of the studies that we, members of the Pak lab and collaborators, carried out from 1998 to 2010 on the functional and physical interactions among several Drosophila phototransduction components. The report includes our studies on the regulations and/or the functions of arrestin II (Arr2), norpA (PLC), inactivation no afterpotential D (INAD), transient receptor potential (TRP), TRP-like (TRPL), inactivation no afterpotential E (INAE), and Porin.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Proteínas do Olho/metabolismo , Transdução de Sinal Luminoso/fisiologia , Animais , Arrestinas/genética , Arrestinas/metabolismo , Proteínas de Drosophila/genética , Proteínas do Olho/genética , Regulação da Expressão Gênica , Canais Iônicos , Canal de Cátion TRPA1 , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Canais de Ânion Dependentes de Voltagem/genética , Canais de Ânion Dependentes de Voltagem/metabolismo
3.
J Neurogenet ; 26(2): 216-37, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22283778

RESUMO

Our objective is to present a comprehensive view of the PDA (prolonged depolarizing afterpotential)-defective Drosophila mutants, nina's and ina's, from the discussion of the PDA and the PDA-based mutant screening strategy to summaries of the knowledge gained through the studies of mutants generated using the strategy. The PDA is a component of the light-evoked photoreceptor potential that is generated when a substantial fraction of rhodopsin is photoconverted to its active form, metarhodopsin. The PDA-based mutant screening strategy was adopted to enhance the efficiency and efficacy of ERG (electroretinogram)-based screening for identifying phototransduction-defective mutants. Using this strategy, two classes of PDA-defective mutants were identified and isolated, nina and ina, each comprising multiple complementation groups. The nina mutants are characterized by allele-dependent reduction in the major rhodopsin, Rh1, whereas the ina mutants display defects in some aspects of functions related to the transduction channel, TRP (transient receptor potential). The signaling proteins that have been identified and elucidated through the studies of nina mutants include the Drosophila opsin protein (NINAE), the chaperone protein for nascent opsin (NINAA), and the multifunctional protein, NINAC, required in multiple steps of the Drosophila phototransduction cascade. Also identified by the nina mutants are some of the key enzymes involved in the biogenesis of the rhodopsin chromophore. As for the ina mutants, they led to the discovery of the scaffold protein, INAD, responsible for the nucleation of the supramolecular signaling complex. Also identified by the ina mutants is one of the key members of the signaling complex, INAC (ePKC), and two other proteins that are likely to be important, though their roles in the signaling cascade have not yet been fully elucidated. In most of these cases, the protein identified is the first member of its class to be so recognized.


Assuntos
Proteínas de Drosophila/deficiência , Proteínas do Olho/metabolismo , Mutação/genética , Células Fotorreceptoras de Invertebrados/fisiologia , Proteínas de Ligação ao Retinol/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Eletrorretinografia , Proteínas do Olho/genética , Testes Genéticos , Proteínas de Ligação ao Retinol/deficiência , Transdução de Sinais/genética
4.
J Neurogenet ; 26(2): 151-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22283835

RESUMO

This is the second of two reviews that include some of the studies we, members of the Pak laboratory and collaborators, did from 2000 to 2010 on the mutants that affect synaptic transmission in the Drosophila visual system. Of the five mutants we discuss, two turned out to also play roles in the larval neuromuscular junction. This review complements the one on phototransduction to give a fairly complete account of what we focused on during the 10-year period, although we also did some studies on photoreceptor degeneration in the early part of the decade. Besides showing the power of using a genetic approach to the study of synaptic transmission, the review contains some unexpected results that illustrate the serendipitous nature of research.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Proteínas do Olho/metabolismo , Transdução de Sinal Luminoso/fisiologia , Sinapses/fisiologia , Vias Visuais/fisiologia , Animais , Canais de Cloreto , Proteínas de Drosophila/genética , Proteínas de Drosophila/história , Proteínas do Olho/genética , Proteínas do Olho/história , Ribonucleoproteínas Nucleares Heterogêneas Grupo F-H , História do Século XX , História do Século XXI , Canais Iônicos , Transdução de Sinal Luminoso/genética , Mutação , Fosfatidilinositol 3-Quinases , Sinapses/genética , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Canal de Cátion TRPA1 , Canais de Cátion TRPC
5.
J Cell Sci ; 123(Pt 19): 3303-15, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20826458

RESUMO

A systematic Drosophila forward genetic screen for photoreceptor synaptic transmission mutants identified no-on-and-no-off transient C (nonC) based on loss of retinal synaptic responses to light stimulation. The cloned gene encodes phosphatidylinositol-3-kinase-like kinase (PIKK) Smg1, a regulatory kinase of the nonsense-mediated decay (NMD) pathway. The Smg proteins act in an mRNA quality control surveillance mechanism to selectively degrade transcripts containing premature stop codons, thereby preventing the translation of truncated proteins with dominant-negative or deleterious gain-of-function activities. At the neuromuscular junction (NMJ) synapse, an extended allelic series of Smg1 mutants show impaired structural architecture, with decreased terminal arbor size, branching and synaptic bouton number. Functionally, loss of Smg1 results in a ~50% reduction in basal neurotransmission strength, as well as progressive transmission fatigue and greatly impaired synaptic vesicle recycling during high-frequency stimulation. Mutation of other NMD pathways genes (Upf2 and Smg6) similarly impairs neurotransmission and synaptic vesicle cycling. These findings suggest that the NMD pathway acts to regulate proper mRNA translation to safeguard synapse morphology and maintain the efficacy of synaptic function.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Células Fotorreceptoras de Invertebrados/metabolismo , Terminações Pré-Sinápticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Proteínas de Drosophila/genética , Teste de Complementação Genética , Testes Genéticos , Transdução de Sinal Luminoso/genética , Morfogênese/genética , Junção Neuromuscular/fisiologia , Células Fotorreceptoras de Invertebrados/patologia , Terminações Pré-Sinápticas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Retina/crescimento & desenvolvimento , Retina/patologia , Deleção de Sequência/genética , Transmissão Sináptica/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/patologia
6.
PLoS One ; 4(8): e6867, 2009 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-19718456

RESUMO

Optic Atrophy 1 (OPA1) is a ubiquitously expressed dynamin-like GTPase in the inner mitochondrial membrane. It plays important roles in mitochondrial fusion, apoptosis, reactive oxygen species (ROS) and ATP production. Mutations of OPA1 result in autosomal dominant optic atrophy (DOA). The molecular mechanisms by which link OPA1 mutations and DOA are not fully understood. Recently, we created a Drosophila model to study the pathogenesis of optic atrophy. Heterozygous mutation of Drosophila OPA1 (dOpa1) by P-element insertion results in no obvious morphological abnormalities, whereas homozygous mutation is embryonic lethal. In eye-specific somatic clones, homozygous mutation of dOpa1 causes rough (mispatterning) and glossy (decreased lens deposition) eye phenotypes in adult Drosophila. In humans, heterozygous mutations in OPA1 have been associated with mitochondrial dysfunction, which is predicted to affect multiple organs. In this study, we demonstrated that heterozygous dOpa1 mutation perturbs the visual function and an ERG profile of the Drosophila compound eye. We independently showed that antioxidants delayed the onset of mutant phenotypes in ERG and improved larval vision function in phototaxis assay. Furthermore, heterozygous dOpa1 mutation also caused decreased heart rate, increased heart arrhythmia, and poor tolerance to stress induced by electrical pacing. However, antioxidants had no effects on the dysfunctional heart of heterozygous dOpa1 mutants. Under stress, heterozygous dOpa1 mutations caused reduced escape response, suggesting abnormal function of the skeletal muscles. Our results suggest that heterozygous mutation of dOpa1 shows organ-specific pathogenesis and is associated with multiple organ abnormalities in an age-dependent and organ-specific manner.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Heterozigoto , Proteínas de Membrana/genética , Mutação , Animais , Antioxidantes/farmacologia , Eletrorretinografia , Modelos Animais , Visão Ocular/efeitos dos fármacos
7.
J Biol Chem ; 284(17): 11100-9, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19254957

RESUMO

Ca(2+) modulates the visual response in both vertebrates and invertebrates. In Drosophila photoreceptors, an increase of cytoplasmic Ca(2+) mimics light adaptation. Little is known regarding the mechanism, however. We explored the role of the sole Drosophila Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) to mediate light adaptation. CaMKII has been implicated in the phosphorylation of arrestin 2 (Arr2). However, the functional significance of Arr2 phosphorylation remains debatable. We identified retinal CaMKII by anti-CaMKII antibodies and by its Ca(2+)-dependent autophosphorylation. Moreover, we show that phosphorylation of CaMKII is greatly enhanced by okadaic acid, and indeed, purified PP2A catalyzes the dephosphorylation of CaMKII. Significantly, we demonstrate that anti-CaMKII antibodies co-immunoprecipitate, and CaMKII fusion proteins pull down the catalytic subunit of PP2A from fly extracts, indicating that PP2A interacts with CaMKII to form a protein complex. To investigate the function of CaMKII in photoreceptors, we show that suppression of CaMKII in transgenic flies affects light adaptation and increases prolonged depolarizing afterpotential amplitude, whereas a reduced PP2A activity brings about reduced prolonged depolarizing afterpotential amplitude. Taken together, we conclude that CaMKII is involved in the negative regulation of the visual response affecting light adaptation, possibly by catalyzing phosphorylation of Arr2. Moreover, the CaMKII activity appears tightly regulated by the co-localized PP2A.


Assuntos
Arrestinas/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Cálcio/metabolismo , Proteínas de Drosophila/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/química , Catálise , Domínio Catalítico , Citosol/metabolismo , Drosophila , Eletrorretinografia/métodos , Heterozigoto , Fenótipo , Fosforilação , Estrutura Terciária de Proteína
8.
Neuron ; 58(6): 884-96, 2008 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-18579079

RESUMO

In Drosophila, a phospholipase C-mediated signaling cascade links photoexcitation of rhodopsin to the opening of the TRP/TRPL channels. A lipid product of the cascade, diacylglycerol (DAG) and its metabolite(s), polyunsaturated fatty acids (PUFAs), have both been proposed as potential excitatory messengers. A crucial enzyme in the understanding of this process is likely to be DAG lipase (DAGL). However, DAGLs that might fulfill this role have not been previously identified in any organism. In this work, the Drosophila DAGL gene, inaE, has been identified from mutants that are defective in photoreceptor responses to light. The inaE-encoded protein isoforms show high sequence similarity to known mammalian DAG lipases, exhibit DAG lipase activity in vitro, and are highly expressed in photoreceptors. Analyses of norpA inaE double mutants and severe inaE mutants show that normal DAGL activity is required for the generation of physiologically meaningful photoreceptor responses.


Assuntos
Proteínas de Drosophila/metabolismo , Lipase Lipoproteica/metabolismo , Células Fotorreceptoras de Invertebrados/fisiologia , Canais de Cátion TRPC/fisiologia , Animais , Drosophila , Proteínas de Drosophila/genética , Ativação Enzimática/fisiologia , Lipase Lipoproteica/genética , Estimulação Luminosa/métodos
9.
J Neurosci ; 28(14): 3668-82, 2008 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-18385325

RESUMO

A systematic forward genetic Drosophila screen for electroretinogram mutants lacking synaptic transients identified the fuseless (fusl) gene, which encodes a predicted eight-pass transmembrane protein in the presynaptic membrane. Null fusl mutants display >75% reduction in evoked synaptic transmission but, conversely, an approximately threefold increase in the frequency and amplitude of spontaneous synaptic vesicle fusion events. These neurotransmission defects are rescued by a wild-type fusl transgene targeted only to the presynaptic cell, demonstrating a strictly presynaptic requirement for Fusl function. Defects in FM dye turnover at the synapse show a severely impaired exo-endo synaptic vesicle cycling pool. Consistently, ultrastructural analyses reveal accumulated vesicles arrested in clustered and docked pools at presynaptic active zones. In the absence of Fusl, calcium-dependent neurotransmitter release is dramatically compromised and there is little enhancement of synaptic efficacy with elevated external Ca(2+) concentrations. These defects are causally linked with severe loss of the Cacophony voltage-gated Ca(2+) channels, which fail to localize normally at presynaptic active zone domains in the absence of Fusl. These data indicate that Fusl regulates assembly of the presynaptic active zone Ca(2+) channel domains required for efficient coupling of the Ca(2+) influx and synaptic vesicle exocytosis during neurotransmission.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Proteínas de Drosophila/fisiologia , Exocitose/fisiologia , Terminações Pré-Sinápticas/metabolismo , Vesículas Sinápticas/fisiologia , Animais , Animais Geneticamente Modificados , Membrana Celular/fisiologia , Drosophila , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Eletrorretinografia/métodos , Embrião não Mamífero , Potenciais Evocados Visuais/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Membrana/fisiologia , Análise em Microsséries , Mutação/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/fisiologia , Junção Neuromuscular/ultraestrutura , Técnicas de Patch-Clamp/métodos , Estimulação Luminosa/métodos , Terminações Pré-Sinápticas/ultraestrutura , Interferência de RNA/fisiologia , Transmissão Sináptica/fisiologia , Visão Ocular/genética , Vias Visuais/anatomia & histologia , Vias Visuais/metabolismo
10.
J Neurosci ; 28(6): 1444-51, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18256265

RESUMO

Drosophila visual signaling, a G-protein-coupled phospholipase Cbeta (PLCbeta)-mediated mechanism, is regulated by eye-protein kinase C (PKC) that promotes light adaptation and fast deactivation, most likely via phosphorylation of inactivation no afterpotential D (INAD) and TRP (transient receptor potential). To reveal the critical phosphatases that dephosphorylate INAD, we used several biochemical analyses and identified protein phosphatase 2A (PP2A) as a candidate. Importantly, the catalytic subunit of PP2A, microtubule star (MTS), is copurified with INAD, and an elevated phosphorylation of INAD by eye-PKC was observed in three mts heterozygotes. To explore whether PP2A (MTS) regulates dephosphorylation of INAD by counteracting eye-PKC [INAC (inactivation no afterpotential C] in vivo, we performed ERG recordings. We discovered that inaC(P209) was semidominant, because inaC(P209) heterozygotes displayed abnormal light adaptation and slow deactivation. Interestingly, the deactivation defect of inaC(P209) heterozygotes was rescued by the mts(XE2258) heterozygous background. In contrast, mts(XE2258) failed to modify the severe deactivation of norpA(P16), indicating that MTS does not modulate NORPA (no receptor potential A) (PLCbeta). Together, our results strongly indicate that dephosphorylation of INAD is catalyzed by PP2A, and a reduction of PP2A can compensate for a partial loss of function in eye-PKC, restoring the fast deactivation kinetics in vivo. We thus propose that the fast deactivation of the visual response is modulated in part by the phosphorylation of INAD.


Assuntos
Proteínas de Drosophila/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Proteína Fosfatase 2/fisiologia , Transdução de Sinais/fisiologia , Percepção Visual/fisiologia , Potenciais de Ação/fisiologia , Adaptação Ocular/fisiologia , Sequência de Aminoácidos , Animais , Catálise , Drosophila , Proteínas de Drosophila/metabolismo , Proteínas do Olho/metabolismo , Proteínas do Olho/fisiologia , Feminino , Masculino , Dados de Sequência Molecular , Fosforilação
11.
Dev Neurobiol ; 67(11): 1533-45, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17525991

RESUMO

Mitochondrial porins, also know as VDACs (voltage-dependent anion channels), play an important role in regulating energy metabolism, apoptosis, and the transport of metabolites across the mitochondrial outer membrane. So far three distinct isoforms of VDAC (VDAC1-3) have been reported in vertebrates, but their functions remain unknown. The annotation database of the Drosophila melanogaster genome sequence has identified four genes (porin, CG17137, CG17139, and CG17140) encoding different isoforms of VDACs. We identified post-translational modifications of PORIN that are specific to D. melanogaster eyes. We also identified the P-element insertion in the porin gene, porin(G2294), that is homozygous viable whereas all the porin mutants previously reported are homozygous lethal at the pupal stage. The mutant does not show any defects in fly morphology, survival, and photoreceptor structure. The mutant, however, produces <10% of the normal level of wild-type (WT) porin transcripts and 16.5% of WT level of the PORIN protein. The P-element insertion affects only the expression of Class I transcript but not Class II transcript of the porin gene. Unlike in WT, the mutant displays an ERG (electroretinogram) that is not maintained during a prolonged light stimulus. The revertant obtained from remobilization of the P-element in the mutant produces the WT level of porin transcripts and PORIN protein, and shows a normal ERG response. Our data suggest that the PORIN protein is important in maintaining a photoreceptor response during prolonged stimulation.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Mitocôndrias/genética , Mutação/genética , Células Fotorreceptoras de Invertebrados/fisiologia , Visão Ocular/genética , Canais de Ânion Dependentes de Voltagem/genética , Potenciais de Ação/genética , Animais , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Eletrorretinografia , Metabolismo Energético/genética , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Estimulação Luminosa , Isoformas de Proteínas/genética , Processamento de Proteína Pós-Traducional/genética , Canais de Ânion Dependentes de Voltagem/metabolismo
12.
J Neurochem ; 89(4): 998-1008, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15140198

RESUMO

A large number of mutants in the norpA gene, which encodes the phospholipase C (PLC) involved in Drosophila phototransduction, is available for the investigation of the effects of specific amino acid substitutions in PLC on biochemical and electrophysiological properties of these mutants. Of the 47 norpA mutants screened for PLC protein content, all but one (H43) displayed drastically decreased amounts of the protein suggesting that almost any mutational alteration has a deleterious effect on the integrity of the protein. Three new amino acids were identified in the catalytic domains X and Y that are important for PLC catalytic activity and the generation of photoreceptor responses (ERG). One of them was found substituted in H43, which showed a low specific PLC activity, a pronounced decrease in ERG sensitivity, and a wild-type-like response termination time. The response termination times obtained from three mutants was found to be approximately inversely proportional to the amount of PLC. In addition, we show that (i) the specific PLC activity is a key factor determining the photoreceptor sensitivity; (ii) the catalytic activity and response termination are separable functions of PLC; and (iii) a mutation in the putative G alpha-interacting C2 domain causes a preferentially strong defect in latency.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Mutação/genética , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Proteínas de Drosophila/deficiência , Drosophila melanogaster/enzimologia , Eletrofisiologia , Eletrorretinografia , Ativação Enzimática/genética , Dados de Sequência Molecular , Fosfolipase C beta , Células Fotorreceptoras de Invertebrados/fisiologia , Tempo de Reação/genética , Homologia de Sequência de Aminoácidos , Fosfolipases Tipo C/deficiência
13.
Recept Channels ; 9(3): 149-67, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12775337

RESUMO

Because almost everything we know about Drosophila phototransduction has come from studies based on genetic approaches, this review begins with a discussion of genetic approaches. We then present a brief overview of Drosophila phototransduction (section on Drosophila phototransduction: an overview) followed by a more detailed treatment of individual components of the transduction machinery (section on Components of the phototransduction machinery). Discussion of transduction mechanisms is presented under three headings: Mechanism(s) of channel excitation, Organization of the transduction proteins, and Regulatory mechanisms in phototransduction. Perhaps the most important unanswered question in this field is the mechanism(s) of activation and regulation of transduction channels. This question is explored in the section entitled Mechanism(s) of channel excitation. Identification of at least two of the proteins discussed was totally unexpected: the rhodopsin chaperone protein, ninaA, and the signal complex scaffold protein, INAD. They are discussed in the sections titled Requirement for a chaperone protein for Rh1 opsin, and: Formation of signaling complexes, respectively. One of the important developments in this field has been the discovery of mammalian homologs of many of the proteins identified in Drosophila. A brief discussion of the most extensively studied of these, the mammalian homologs of light-activated channel protein, trp, is presented in the section on Mammalian Homologs of trp. We conclude the review with Perspective, a brief look at the current status and the future outlook of the field.


Assuntos
Drosophila melanogaster/genética , Transdução de Sinais , Visão Ocular/fisiologia , Animais , Drosophila melanogaster/fisiologia , Potenciais Evocados Visuais
14.
J Biol Chem ; 277(44): 42113-20, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12196539

RESUMO

By screening Drosophila mutants that are potentially defective in synaptic transmission between photoreceptors and their target laminar neurons, L1/L2, (lack of electroretinogram on/off transients), we identified ort as a candidate gene encoding a histamine receptor subunit on L1/L2. We provide evidence that the ort gene corresponds to CG7411 (referred to as hclA), identified in the Drosophila genome data base, by P-element-mediated germ line rescue of the ort phenotype using cloned hclA cDNA and by showing that several ort mutants exhibit alterations in hclA regulatory or coding sequences and/or allele-dependent reductions in hclA transcript levels. Other workers have shown that hclA, when expressed in Xenopus oocytes, forms histamine-sensitive chloride channels. However, the connection between these chloride channels and photoreceptor synaptic transmission was not established. We show unequivocally that hclA-encoded channels are the channels required in photoreceptor synaptic transmission by 1) establishing the identity between hclA and ort and 2) showing that ort mutants are defective in photoreceptor synaptic transmission. Moreover, the present work shows that this function of the HCLA (ORT) protein is its native function in vivo.


Assuntos
Canais de Cloreto/genética , Drosophila/genética , Genes de Insetos , Células Fotorreceptoras de Invertebrados/fisiologia , Receptores Histamínicos/genética , Transmissão Sináptica , Sequência de Aminoácidos , Animais , Northern Blotting , Canais de Cloreto/fisiologia , DNA Complementar/isolamento & purificação , Dados de Sequência Molecular , Mutação , Subunidades Proteicas , RNA Mensageiro/análise , Receptores Histamínicos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...