Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 145(7): 1088-101, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21703451

RESUMO

INAD is a scaffolding protein that regulates signaling in Drosophila photoreceptors. One of its PDZ domains, PDZ5, cycles between reduced and oxidized forms in response to light, but it is unclear how light affects its redox potential. Through biochemical and structural studies, we show that the redox potential of PDZ5 is allosterically regulated by its interaction with another INAD domain, PDZ4. Whereas isolated PDZ5 is stable in the oxidized state, formation of a PDZ45 "supramodule" locks PDZ5 in the reduced state by raising the redox potential of its Cys606/Cys645 disulfide bond by ∼330 mV. Acidification, potentially mediated via light and PLCß-mediated hydrolysis of PIP(2), disrupts the interaction between PDZ4 and PDZ5, leading to PDZ5 oxidation and dissociation from the TRP Ca(2+) channel, a key component of fly visual signaling. These results show that scaffolding proteins can actively modulate the intrinsic redox potentials of their disulfide bonds to exert regulatory roles in signaling.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas do Olho/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Drosophila/química , Olho/metabolismo , Proteínas do Olho/química , Modelos Moleculares , Oxirredução , Domínios PDZ , Células Fotorreceptoras de Invertebrados/metabolismo , Transdução de Sinais
2.
Proc Natl Acad Sci U S A ; 119(12): e2109717119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35298337

RESUMO

SignificanceTo move efficiently, animals must continuously work out their x,y,z positions with respect to real-world objects, and many animals have a pair of eyes to achieve this. How photoreceptors actively sample the eyes' optical image disparity is not understood because this fundamental information-limiting step has not been investigated in vivo over the eyes' whole sampling matrix. This integrative multiscale study will advance our current understanding of stereopsis from static image disparity comparison to a morphodynamic active sampling theory. It shows how photomechanical photoreceptor microsaccades enable Drosophila superresolution three-dimensional vision and proposes neural computations for accurately predicting these flies' depth-perception dynamics, limits, and visual behaviors.


Assuntos
Percepção de Profundidade , Drosophila , Animais , Olho , Disparidade Visual , Visão Ocular
3.
Cell ; 139(2): 246-64, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19837030

RESUMO

Seeing begins in the photoreceptors, where light is absorbed and signaled to the nervous system. Throughout the animal kingdom, photoreceptors are diverse in design and purpose. Nonetheless, phototransduction-the mechanism by which absorbed photons are converted into an electrical response-is highly conserved and based almost exclusively on a single class of photoproteins, the opsins. In this Review, we survey the G protein-coupled signaling cascades downstream from opsins in photoreceptors across vertebrate and invertebrate species, noting their similarities as well as differences.


Assuntos
Transdução de Sinal Luminoso , Receptores Acoplados a Proteínas G/metabolismo , Animais , Humanos , Células Fotorreceptoras de Invertebrados/metabolismo , Células Fotorreceptoras de Vertebrados
4.
J Neurosci ; 40(16): 3152-3164, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32156830

RESUMO

Phototransduction in Drosophila is mediated by phospholipase C (PLC) and Ca2+-permeable TRP channels, but the function of endoplasmic reticulum (ER) Ca2+ stores in this important model for Ca2+ signaling remains obscure. We therefore expressed a low affinity Ca2+ indicator (ER-GCaMP6-150) in the ER, and measured its fluorescence both in dissociated ommatidia and in vivo from intact flies of both sexes. Blue excitation light induced a rapid (tau ∼0.8 s), PLC-dependent decrease in fluorescence, representing depletion of ER Ca2+ stores, followed by a slower decay, typically reaching ∼50% of initial dark-adapted levels, with significant depletion occurring under natural levels of illumination. The ER stores refilled in the dark within 100-200 s. Both rapid and slow store depletion were largely unaffected in InsP3 receptor mutants, but were much reduced in trp mutants. Strikingly, rapid (but not slow) depletion of ER stores was blocked by removing external Na+ and in mutants of the Na+/Ca2+ exchanger, CalX, which we immuno-localized to ER membranes in addition to its established localization in the plasma membrane. Conversely, overexpression of calx greatly enhanced rapid depletion. These results indicate that rapid store depletion is mediated by Na+/Ca2+ exchange across the ER membrane induced by Na+ influx via the light-sensitive channels. Although too slow to be involved in channel activation, this Na+/Ca2+ exchange-dependent release explains the decades-old observation of a light-induced rise in cytosolic Ca2+ in photoreceptors exposed to Ca2+-free solutions.SIGNIFICANCE STATEMENT Phototransduction in Drosophila is mediated by phospholipase C, which activates TRP cation channels by an unknown mechanism. Despite much speculation, it is unknown whether endoplasmic reticulum (ER) Ca2+ stores play any role. We therefore engineered flies expressing a genetically encoded Ca2+ indicator in the photoreceptor ER. Although NCX Na+/Ca2+ exchangers are classically believed to operate only at the plasma membrane, we demonstrate a rapid light-induced depletion of ER Ca2+ stores mediated by Na+/Ca2+ exchange across the ER membrane. This NCX-dependent release was too slow to be involved in channel activation, but explains the decades-old observation of a light-induced rise in cytosolic Ca2+ in photoreceptors bathed in Ca2+-free solutions.


Assuntos
Antiporters/metabolismo , Cálcio/metabolismo , Proteínas de Drosophila/metabolismo , Retículo Endoplasmático/metabolismo , Transdução de Sinal Luminoso/fisiologia , Células Fotorreceptoras de Invertebrados/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Animais , Animais Geneticamente Modificados , Antiporters/genética , Sinalização do Cálcio/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Masculino , Trocador de Sódio e Cálcio/genética
5.
J Neurosci ; 39(36): 7132-7154, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31350259

RESUMO

Ca2+-activated K+ channels (BK and SK) are ubiquitous in synaptic circuits, but their role in network adaptation and sensory perception remains largely unknown. Using electrophysiological and behavioral assays and biophysical modeling, we discover how visual information transfer in mutants lacking the BK channel (dSlo- ), SK channel (dSK- ), or both (dSK- ;; dSlo- ) is shaped in the female fruit fly (Drosophila melanogaster) R1-R6 photoreceptor-LMC circuits (R-LMC-R system) through synaptic feedforward-feedback interactions and reduced R1-R6 Shaker and Shab K+ conductances. This homeostatic compensation is specific for each mutant, leading to distinctive adaptive dynamics. We show how these dynamics inescapably increase the energy cost of information and promote the mutants' distorted motion perception, determining the true price and limits of chronic homeostatic compensation in an in vivo genetic animal model. These results reveal why Ca2+-activated K+ channels reduce network excitability (energetics), improving neural adaptability for transmitting and perceiving sensory information.SIGNIFICANCE STATEMENT In this study, we directly link in vivo and ex vivo experiments with detailed stochastically operating biophysical models to extract new mechanistic knowledge of how Drosophila photoreceptor-interneuron-photoreceptor (R-LMC-R) circuitry homeostatically retains its information sampling and transmission capacity against chronic perturbations in its ion-channel composition, and what is the cost of this compensation and its impact on optomotor behavior. We anticipate that this novel approach will provide a useful template to other model organisms and computational neuroscience, in general, in dissecting fundamental mechanisms of homeostatic compensation and deepening our understanding of how biological neural networks work.


Assuntos
Retroalimentação Fisiológica , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Potenciais Sinápticos , Percepção Visual , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Interneurônios/metabolismo , Interneurônios/fisiologia , Modelos Neurológicos , Células Fotorreceptoras de Invertebrados/fisiologia , Canais de Potássio Shab/metabolismo , Superfamília Shaker de Canais de Potássio/metabolismo , Vias Visuais/metabolismo , Vias Visuais/fisiologia
7.
J Cell Sci ; 131(8)2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29567856

RESUMO

Phototransduction in Drosophila is mediated by phospholipase C-dependent hydrolysis of PIP2-, and is an important model for phosphoinositide signalling. Although generally assumed to operate by generic machinery conserved from yeast to mammals, some key elements of the phosphoinositide cycle have yet to be identified in Drosophila photoreceptors. Here, we used transgenic flies expressing fluorescently tagged probes (P4M and TbR332H), which allow in vivo quantitative measurements of PI4P and PIP2 dynamics in photoreceptors of intact living flies. Using mutants and RNA interference for candidate genes potentially involved in phosphoinositide turnover, we identified Drosophila PI4KIIIα (CG10260) as the PI4-kinase responsible for PI4P synthesis in the photoreceptor membrane. Our results also indicate that PI4KIIIα activity requires rbo (the Drosophila orthologue of Efr3) and CG8325 (orthologue of YPP1), both of which are implicated as scaffolding proteins necessary for PI4KIIIα activity in yeast and mammals. However, our evidence indicates that the recently reported central role of dPIP5K59B (CG3682) in PIP2 synthesis in the rhabdomeres should be re-evaluated; although PIP2 resynthesis was suppressed by RNAi directed against dPIP5K59B, little or no defect was detected in a reportedly null mutant (dPIP5K18 ).


Assuntos
Fosfatidilinositóis/genética , Células Fotorreceptoras/metabolismo , Animais , Drosophila , Fosfatidilinositóis/metabolismo
8.
J Cell Sci ; 128(23): 4328-40, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26483384

RESUMO

In order to monitor phosphoinositide turnover during phospholipase C (PLC)-mediated Drosophila phototransduction, fluorescently tagged lipid probes were expressed in photoreceptors and imaged both in dissociated cells, and in eyes of intact living flies. Of six probes tested, Tb(R332H) (a mutant of the Tubby protein pleckstrin homology domain) was judged the best reporter for phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P2], and the P4M domain from Legionella SidM for phosphatidylinositol 4-phosphate (PtdIns4P). Using accurately calibrated illumination, we found that only ∼50% of PtdIns(4,5)P2 and very little PtdIns4P were depleted by full daylight intensities in wild-type flies, but both were severely depleted by ∼100-fold dimmer intensities in mutants lacking Ca(2+)-permeable transient receptor potential (TRP) channels or protein kinase C (PKC). Resynthesis of PtdIns4P (t½ ∼12 s) was faster than PtdIns(4,5)P2 (t½ ∼40 s), but both were greatly slowed in mutants of DAG kinase (rdgA) or PtdIns transfer protein (rdgB). The results indicate that Ca(2+)- and PKC-dependent inhibition of PLC is required for enabling photoreceptors to maintain phosphoinositide levels despite high rates of hydrolysis by PLC, and suggest that phosphorylation of PtdIns4P to PtdIns(4,5)P2 is the rate-limiting step of the cycle.


Assuntos
Fosfatidilinositol 4,5-Difosfato/metabolismo , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/metabolismo , Animais , Cálcio/metabolismo , Diacilglicerol Quinase/genética , Diacilglicerol Quinase/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfatidilinositol 4,5-Difosfato/genética , Proteína Quinase C/genética , Proteína Quinase C/metabolismo
9.
J Neurosci ; 35(6): 2731-46, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25673862

RESUMO

Drosophila phototransduction is mediated via a G-protein-coupled PLC cascade. Recent evidence, including the demonstration that light evokes rapid contractions of the photoreceptors, suggested that the light-sensitive channels (TRP and TRPL) may be mechanically gated, together with protons released by PLC-mediated PIP2 hydrolysis. If mechanical gating is involved we predicted that the response to light should be influenced by altering the physical properties of the membrane. To achieve this, we used diet to manipulate the degree of saturation of membrane phospholipids. In flies reared on a yeast diet, lacking polyunsaturated fatty acids (PUFAs), mass spectrometry showed that the proportion of polyunsaturated phospholipids was sevenfold reduced (from 38 to ∼5%) but rescued by adding a single species of PUFA (linolenic or linoleic acid) to the diet. Photoreceptors from yeast-reared flies showed a 2- to 3-fold increase in latency and time to peak of the light response, without affecting quantum bump waveform. In the absence of Ca(2+) influx or in trp mutants expressing only TRPL channels, sensitivity to light was reduced up to ∼10-fold by the yeast diet, and essentially abolished in hypomorphic G-protein mutants (Gαq). PLC activity appeared little affected by the yeast diet; however, light-induced contractions measured by atomic force microscopy or the activation of ectopic mechanosensitive gramicidin channels were also slowed ∼2-fold. The results are consistent with mechanosensitive gating and provide a striking example of how dietary fatty acids can profoundly influence sensory performance in a classical G-protein-coupled signaling cascade.


Assuntos
Membrana Celular/fisiologia , Drosophila melanogaster/fisiologia , Transdução de Sinal Luminoso/fisiologia , Fosfolipídeos/fisiologia , Animais , Membrana Celular/metabolismo , Dieta , Ativação do Canal Iônico/fisiologia , Luz , Metabolismo dos Lipídeos/fisiologia , Fosfolipídeos/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Rodopsina/metabolismo , Razão Sinal-Ruído , Trocador de Sódio e Cálcio/metabolismo , Fosfolipases Tipo C/metabolismo
10.
J Cell Sci ; 126(Pt 5): 1247-59, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23378018

RESUMO

The prototypical transient receptor potential (TRP) channel is the major light-sensitive, and Ca(2+)-permeable channel in the microvillar photoreceptors of Drosophila. TRP channels are activated following hydrolysis of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] by the key effector enzyme phospholipase C (PLC). Mutants lacking TRP channels undergo light-dependent retinal degeneration, as a consequence of the reduced Ca(2+) influx. It has been proposed that degeneration is caused by defects in the Ca(2+)-dependent visual pigment cycle, which result in accumulation of toxic phosphorylated metarhodopsin-arrestin complexes (MPP-Arr2). Here we show that two interventions, which prevent accumulation of MPP-Arr2, namely rearing under red light or eliminating the C-terminal rhodopsin phosphorylation sites, failed to rescue degeneration in trp mutants. Instead, degeneration in trp mutants reared under red light was rescued by mutation of PLC. Degeneration correlated closely with the light-induced depletion of PtdIns(4,5)P2 that occurs in trp mutants due to failure of Ca(2+)-dependent inhibition of PLC. Severe retinal degeneration was also induced in the dark in otherwise wild-type flies by overexpression of a bacterial PtdInsPn phosphatase (SigD) to deplete PtdIns(4,5)P2. In degenerating trp photoreceptors, phosphorylated Moesin, a PtdIns(4,5)P2-regulated membrane-cytoskeleton linker essential for normal microvillar morphology, was found to delocalize from the rhabdomere and there was extensive microvillar actin depolymerisation. The results suggest that compromised light-induced Ca(2+) influx, due to loss of TRP channels, leads to PtdIns(4,5)P2 depletion, resulting in dephosphorylation of Moesin, actin depolymerisation and disintegration of photoreceptor structure.


Assuntos
Fosfatidilinositol 4,5-Difosfato/metabolismo , Degeneração Retiniana/fisiopatologia , Actinas/genética , Actinas/metabolismo , Animais , Drosophila , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Degeneração Retiniana/genética , Rodopsina/metabolismo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo
11.
Handb Exp Pharmacol ; 223: 795-826, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24961970

RESUMO

The Drosophila "transient receptor potential" channel is the prototypical TRP channel, belonging to and defining the TRPC subfamily. Together with a second TRPC channel, trp-like (TRPL), TRP mediates the transducer current in the fly's photoreceptors. TRP and TRPL are also implicated in olfaction and Malpighian tubule function. In photoreceptors, TRP and TRPL are localised in the ~30,000 packed microvilli that form the photosensitive "rhabdomere"-a light-guiding rod, housing rhodopsin and the rest of the phototransduction machinery. TRP (but not TRPL) is assembled into multimolecular signalling complexes by a PDZ-domain scaffolding protein (INAD). TRPL (but not TRP) undergoes light-regulated translocation between cell body and rhabdomere. TRP and TRPL are also found in photoreceptor synapses where they may play a role in synaptic transmission. Like other TRPC channels, TRP and TRPL are activated by a G protein-coupled phospholipase C (PLCß4) cascade. Although still debated, recent evidence indicates the channels can be activated by a combination of PIP2 depletion and protons released by the PLC reaction. PIP2 depletion may act mechanically as membrane area is reduced by cleavage of PIP2's bulky inositol headgroup. TRP, which dominates the light-sensitive current, is Ca(2+) selective (P Ca:P Cs >50:1), whilst TRPL has a modest Ca(2+) permeability (P Ca:P Cs ~5:1). Ca(2+) influx via the channels has profound positive and negative feedback roles, required for the rapid response kinetics, with Ca(2+) rapidly facilitating TRP (but not TRPL) and also inhibiting both channels. In trp mutants, stimulation by light results in rapid depletion of microvillar PIP2 due to lack of Ca(2+) influx required to inhibit PLC. This accounts for the "transient receptor potential" phenotype that gives the family its name and, over a period of days, leads to light-dependent retinal degeneration. Gain-of-function trp mutants with uncontrolled Ca(2+) influx also undergo retinal degeneration due to Ca(2+) cytotoxicity. In vertebrate retina, mice knockout studies suggest that TRPC6 and TRPC7 mediate a PLCß4-activated transducer current in intrinsically photosensitive retinal ganglion cells, expressing melanopsin. TRPA1 has been implicated as a "photo-sensing" TRP channel in human melanocytes and light-sensitive neurons in the body wall of Drosophila.


Assuntos
Células Fotorreceptoras/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Cálcio/metabolismo , Drosophila/fisiologia , Proteínas de Drosophila/fisiologia , Humanos , Canais de Potencial de Receptor Transitório/química , Canais de Potencial de Receptor Transitório/genética
12.
Biophys J ; 104(9): 1905-16, 2013 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-23663833

RESUMO

Light responses in Drosophila photoreceptors are mediated by two Ca(2+) permeable cation channels, transient receptor potential (TRP) and TRP-like (TRPL). Although Ca(2+) influx via these channels is critical for amplification, inactivation, and light adaptation, the fractional contribution of Ca(2+) to the currents (Pf) has not been measured. We describe a slow (τ ∼ 350 ms) tail current in voltage-clamped light responses and show that it is mediated by electrogenic Na(+)/Ca(2+) exchange. Assuming a 3Na:1Ca stoichiometry, we derive empirical estimates of Pf by comparing the charge integrals of the exchanger and light-induced currents. For TRPL channels, Pf was ∼17% as predicted by Goldman-Hodgkin-Katz (GHK) theory. Pf for TRP (29%) and wild-type flies (26%) was higher, but lower than the GHK prediction (45% and 42%). As predicted by GHK theory, Pf for both channels increased with extracellular [Ca(2+)], and was largely independent of voltage between -100 and -30 mV. A model incorporating intra- and extracellular geometry, ion permeation, diffusion, extrusion, and buffering suggested that the deviation from GHK predictions was largely accounted for by extracellular ionic depletion during the light-induced currents, and the time course of the Na(+)/Ca(2+) exchange current could be used to obtain estimates of cellular Ca(2+) buffering capacities.


Assuntos
Potenciais de Ação , Cálcio/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Antiporters/genética , Antiporters/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Transporte de Íons , Mutação , Estimulação Luminosa , Células Fotorreceptoras de Invertebrados/fisiologia , Sódio/metabolismo , Canais de Potencial de Receptor Transitório/genética
13.
J Neurosci ; 32(27): 9205-16, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22764229

RESUMO

Upon illumination several phototransduction proteins translocate between cell body and photosensory compartments. In Drosophila photoreceptors arrestin (Arr2) translocates from cell body to the microvillar rhabdomere down a diffusion gradient created by binding of Arr2 to photo-isomerized metarhodopsin. Translocation is profoundly slowed in mutants of key phototransduction proteins including phospholipase C (PLC) and the Ca(2+)-permeable transient receptor potential channel (TRP), but how the phototransduction cascade accelerates Arr2 translocation is unknown. Using real-time fluorescent imaging of Arr2-green fluorescent protein translocation in dissociated ommatidia, we show that translocation is profoundly slowed in Ca(2+)-free solutions. Conversely, in a blind PLC mutant with ∼100-fold slower translocation, rapid translocation was rescued by the Ca(2+) ionophore, ionomycin. In mutants lacking NINAC (calmodulin [CaM] binding myosin III) in the cell body, translocation remained rapid even in Ca(2+)-free solutions. Immunolabeling revealed that Arr2 in the cell body colocalized with NINAC in the dark. In intact eyes, the impaired translocation found in trp mutants was rescued in ninaC;trp double mutants. Nevertheless, translocation following prolonged dark adaptation was significantly slower in ninaC mutants, than in wild type: a difference that was reflected in the slow decay of the electroretinogram. The results suggest that cytosolic NINAC is a Ca(2+)-dependent binding target for Arr2, which protects Arr2 from immobilization by a second potential sink that sequesters and releases arrestin on a much slower timescale. We propose that rapid Ca(2+)/CaM-dependent release of Arr2 from NINAC upon Ca(2+) influx accounts for the acceleration of translocation by phototransduction.


Assuntos
Arrestinas/metabolismo , Cálcio/fisiologia , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Cadeias Pesadas de Miosina/deficiência , Miosina Tipo III/fisiologia , Células Fotorreceptoras de Invertebrados/metabolismo , Visão Ocular/fisiologia , Animais , Animais Geneticamente Modificados , Arrestinas/genética , Sinalização do Cálcio/genética , Sinalização do Cálcio/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Proteínas do Olho/genética , Feminino , Masculino , Cadeias Pesadas de Miosina/genética , Miosina Tipo III/genética , Células Fotorreceptoras de Invertebrados/citologia , Transporte Proteico/fisiologia
14.
J Neurophysiol ; 109(8): 2044-55, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23365183

RESUMO

Absolute visual thresholds are limited by "dark noise," which in Drosophila photoreceptors is dominated by brief (∼10 ms), small (∼2 pA) inward current events, occurring at ∼2/s, believed to reflect spontaneous G protein activations. These dark events were increased in rate and amplitude by a point mutation in myosin III (NINAC), which disrupts its interaction with the scaffolding protein, INAD. This phenotype mimics that previously described in null mutants of ninaC (no inactivation no afterpotential; encoding myosin III) and an associated protein, retinophilin (rtp). Dark noise was similarly increased in heterozygote mutants of diacylglycerol kinase (rdgA/+). Dark noise in ninaC, rtp, and rdgA/+ mutants was greatly suppressed by mutations of the Gq α-subunit (Gαq) and the major light-sensitive channel (trp) but not rhodopsin. ninaC, rtp, and rdgA/+ mutations also all facilitated residual light responses in Gαq and PLC hypomorphs. Raising cytosolic Ca(2+) in the submicromolar range increased dark noise, facilitated activation of transient receptor potential (TRP) channels by exogenous agonist, and again facilitated light responses in Gαq hypomorphs. Our results indicate that RTP, NINAC, INAD, and diacylglycerol kinase, together with a Ca(2+)-dependent threshold, share common roles in suppressing dark noise and regulating quantum bump generation; consequently, most spontaneous G protein activations fail to generate dark events under normal conditions. By contrast, quantum bump generation is reliable but delayed until sufficient G proteins and PLC are activated to overcome threshold, thereby ensuring generation of full-size bumps with high quantum efficiency.


Assuntos
Potenciais de Ação , Drosophila/fisiologia , Células Fotorreceptoras de Invertebrados/fisiologia , Animais , Cálcio/metabolismo , Diacilglicerol Quinase/genética , Diacilglicerol Quinase/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Heterozigoto , Mutação , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Limiar Sensorial , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
15.
J Neurosci ; 31(39): 13897-910, 2011 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-21957252

RESUMO

The contribution of the SK (small-conductance calcium-activated potassium) channel to neuronal functions in complex circuits underlying sensory processing and behavior is largely unknown in the absence of suitable animal models. Here, we generated a Drosophila line that lacks the single highly conserved SK gene in its genome (dSK). In R1-R6 photoreceptors, dSK encodes a slow Ca²âº-activated K(+) current similar to its mammalian counterparts. Compared with wild-type, dSK(-) photoreceptors and interneurons showed accelerated oscillatory responses and adaptation. These enhanced kinetics were accompanied with more depolarized dSK(-) photoreceptors axons, assigning a role for dSK in network gain control during light-to-dark transitions. However, compensatory network adaptation, through increasing activity between synaptic neighbors, overcame many detriments of missing dSK current enabling dSK(-) photoreceptors to maintain normal information transfer rates to naturalistic stimuli. While demonstrating important functional roles for dSK channel in the visual circuitry, these results also clarify how homeostatically balanced network functions can compensate missing or faulty ion channels.


Assuntos
Proteínas de Drosophila/fisiologia , Rede Nervosa/fisiologia , Células Fotorreceptoras/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Vias Visuais/fisiologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Masculino , Dados de Sequência Molecular , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
16.
J Neurosci ; 30(4): 1238-49, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20107052

RESUMO

Photoreceptor cells achieve high sensitivity, reliably detecting single photons, while limiting the spontaneous activation events responsible for dark noise. We used proteomic, genetic, and electrophysiological approaches to characterize Retinophilin (RTP) (CG10233) in Drosophila photoreceptors and establish its involvement in dark-noise suppression. RTP possesses membrane occupation and recognition nexus (MORN) motifs, a structure shared with mammalian junctophilins and other membrane-associated proteins found within excitable cells. We show the MORN repeats, and both the N- and C-terminal domains, are required for RTP localization in the microvillar light-gathering organelle, the rhabdomere. RTP exists in multiple phosphorylated isoforms under dark conditions and is dephosphorylated by light exposure. An RTP deletion mutant exhibits a high rate of spontaneous membrane depolarization events in dark conditions but retains the normal kinetics of the light response. Photoreceptors lacking neither inactivation nor afterpotential C (NINAC) myosin III, a motor protein/kinase, also display a similar dark-noise phenotype as the RTP deletion. We show that NINAC mutants are depleted for RTP. These results suggest the increase in dark noise in NINAC mutants is attributable to lack of RTP and, furthermore, defines a novel role for NINAC in the rhabdomere. We propose that RTP is a light-regulated phosphoprotein that organizes rhabdomeric components to suppress random activation of the phototransduction cascade and thus increases the signaling fidelity of dark-adapted photoreceptors.


Assuntos
Adaptação à Escuridão/efeitos da radiação , Proteínas de Drosophila/metabolismo , Proteínas do Olho/metabolismo , Olho/metabolismo , Fosfoproteínas/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Visão Ocular/fisiologia , Adaptação Ocular/fisiologia , Adaptação Ocular/efeitos da radiação , Motivos de Aminoácidos/fisiologia , Animais , Animais Geneticamente Modificados , Adaptação à Escuridão/fisiologia , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster , Olho/ultraestrutura , Proteínas do Olho/química , Proteínas do Olho/genética , Luz , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/efeitos da radiação , Microvilosidades/metabolismo , Microvilosidades/efeitos da radiação , Microvilosidades/ultraestrutura , Mutação/genética , Fosfoproteínas/genética , Estimulação Luminosa , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/efeitos da radiação , Células Fotorreceptoras/ultraestrutura , Células Fotorreceptoras de Invertebrados/efeitos da radiação , Células Fotorreceptoras de Invertebrados/ultraestrutura , Estrutura Terciária de Proteína/fisiologia , Estrutura Terciária de Proteína/efeitos da radiação , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação
17.
Pflugers Arch ; 461(5): 493-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21286746

RESUMO

The history of the discovery of the transient receptor potential (TRP) cation channel superfamily began in 1969 with Cosens and Manning's isolation of the Drosophila transient receptor potential mutant, in which the photoreceptor response decays during continuous illumination. Early studies from Minke found that the elementary light response was unaffected in trp mutants, and he attributed the defect to an intermediate stage of phototransduction. Montell and Rubin cloned the trp gene in 1989: they recognised it as a transmembrane protein, but also concluded that it did not encode the light-sensitive channels. In 1991, Minke and Selinger proposed that TRP represented a Ca2+ transporter required for refilling intracellular InsP3-sensitive Ca2+ stores, in turn required for activation of the light-sensitive channels. Also in 1991, after developing a photoreceptor patch clamp preparation, I showed that the light-sensitive channels themselves were highly permeable to Ca2+, questioning the need for such a dedicated Ca2+ transporter. In 1992, in collaboration with Minke, I resolved this paradox by showing there were two classes of light-sensitive channels, one highly Ca2+ permeable and eliminated in trp mutants. This represented the first and compelling evidence that TRP represented a light-sensitive channel and was supported by the cloning of the second light-sensitive channel, TRPL, by Kelly's lab. Three years later, in 1995, the labs of Montell and Birnbaumer independently cloned TRPC1, the first of 29 vertebrate TRP isoforms distributed amongst seven subfamilies.


Assuntos
Canais de Cálcio/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Cálcio/metabolismo , Drosophila , Luz , Canais de Potencial de Receptor Transitório/genética
19.
Artigo em Inglês | MEDLINE | ID: mdl-21046112

RESUMO

The visual pigments of most invertebrate photoreceptors have two thermostable photo-interconvertible states, the ground state rhodopsin and photo-activated metarhodopsin, which triggers the phototransduction cascade until it binds arrestin. The ratio of the two states in photoequilibrium is determined by their absorbance spectra and the effective spectral distribution of illumination. Calculations indicate that metarhodopsin levels in fly photoreceptors are maintained below ~35% in normal diurnal environments, due to the combination of a blue-green rhodopsin, an orange-absorbing metarhodopsin and red transparent screening pigments. Slow metarhodopsin degradation and rhodopsin regeneration processes further subserve visual pigment maintenance. In most insect eyes, where the majority of photoreceptors have green-absorbing rhodopsins and blue-absorbing metarhodopsins, natural illuminants are predicted to create metarhodopsin levels greater than 60% at high intensities. However, fast metarhodopsin decay and rhodopsin regeneration also play an important role in controlling metarhodopsin in green receptors, resulting in a high rhodopsin content at low light intensities and a reduced overall visual pigment content in bright light. A simple model for the visual pigment-arrestin cycle is used to illustrate the dependence of the visual pigment population states on light intensity, arrestin levels and pigment turnover.


Assuntos
Arrestina/fisiologia , Olho Composto de Artrópodes/metabolismo , Dípteros/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Rodopsina/metabolismo , Visão Ocular/fisiologia , Animais , Olho Composto de Artrópodes/citologia , Olho Composto de Artrópodes/efeitos da radiação , Adaptação à Escuridão/fisiologia , Dípteros/citologia , Luz , Microvilosidades/metabolismo , Microvilosidades/efeitos da radiação , Estimulação Luminosa/métodos , Fotoperíodo , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/efeitos da radiação
20.
Structure ; 29(4): 330-344.e4, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33326749

RESUMO

Drosophila TRP is a calcium-permeable cation channel essential for fly visual signal transduction. During phototransduction, Ca2+ mediates both positive and negative feedback regulation on TRP channel activity, possibly via binding to calmodulin (CaM). However, the molecular mechanism underlying Ca2+ modulated CaM/TRP interaction is poorly understood. Here, we discover an unexpected, Ca2+-dependent binding mode between CaM and TRP. The TRP tail contains two CaM binding sites (CBS1 and CBS2) separated by an ∼70-residue linker. CBS1 binds to the CaM N-lobe and CBS2 recognizes the CaM C-lobe. Structural studies reveal the lobe-specific binding of CaM to CBS1&2. Mutations introduced in both CBS1 and CBS2 eliminated CaM binding in full-length TRP, but surprisingly had no effect on the response to light under physiological conditions, suggesting alternative mechanisms governing Ca2+-mediated feedback on the channel activity. Finally, we discover that TRPC4, the closest mammalian paralog of Drosophila TRP, adopts a similar CaM binding mode.


Assuntos
Calmodulina/química , Proteínas de Drosophila/química , Canais de Potencial de Receptor Transitório/química , Animais , Sítios de Ligação , Cálcio/metabolismo , Calmodulina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Células HEK293 , Humanos , Camundongos , Mutação , Ligação Proteica , Canais de Cátion TRPC/química , Canais de Cátion TRPC/metabolismo , Canais de Potencial de Receptor Transitório/genética , Canais de Potencial de Receptor Transitório/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA