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.
Cells ; 9(7)2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-32668787

RESUMO

Anomalies in constitutive calcium entry (CCE) have been commonly attributed to cell dysfunction in pathological conditions such as cancer. Calcium influxes of this type rely on channels, such as transient receptor potential (TRP) channels, to be constitutively opened and strongly depend on membrane potential and a calcium driving force. We developed an optogenetic approach based on the expression of the halorhodopsin chloride pump to study CCE in non-excitable cells. Using C2C12 cells, we found that halorhodopsin can be used to achieve a finely tuned control of membrane polarization. Escalating the membrane polarization by incremental changes in light led to a concomitant increase in CCE through transient receptor potential vanilloid 2 (TRPV2) channels. Moreover, light-induced calcium entry through TRPV2 channels promoted cell migration. Our study shows for the first time that by modulating CCE and related physiological responses, such as cell motility, halorhodopsin serves as a potentially powerful tool that could open new avenues for the study of CCE and associated cellular behaviors.


Assuntos
Cálcio/metabolismo , Movimento Celular , Potenciais da Membrana , Optogenética , Animais , Canais de Cálcio/metabolismo , Linhagem Celular , Movimento Celular/efeitos da radiação , Halorrodopsinas/metabolismo , Humanos , Luz , Potenciais da Membrana/efeitos da radiação , Camundongos , Mioblastos/metabolismo , Mioblastos/efeitos da radiação , Canais de Cátion TRPV/metabolismo
2.
Stem Cells Dev ; 29(4): 187-197, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31559914

RESUMO

Although research involving traumatic brain injury (TBI) has traditionally focused on the acute clinical manifestations, new studies provide evidence for chronic and progressive neurological sequelae associated with TBI, highlighting the risk of persistent, and sometimes life-long, consequences for affected patients. Several treatment modalities to date have demonstrated efficacy in experimental models. However, there is currently no effective treatment to improve neural structure repair and functional recovery of TBI patients. Optogenetics represents a potential molecular tool for neuromodulation and monitoring cellular activity with unprecedented spatial resolution and millisecond temporal precision. In this review, we discuss the conceptual background and preclinical evidence of optogenetics for neuromodulation, and translational applications for TBI treatment are considered.


Assuntos
Proteínas Arqueais/genética , Lesões Encefálicas Traumáticas/genética , Channelrhodopsins/genética , Halorrodopsinas/genética , Terapia de Alvo Molecular/métodos , Optogenética/métodos , Animais , Animais Geneticamente Modificados , Proteínas Arqueais/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/terapia , Channelrhodopsins/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Halorrodopsinas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Optogenética/tendências , Transmissão Sináptica , Pesquisa Translacional Biomédica
3.
Nat Biomed Eng ; 2(7): 485-496, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30948823

RESUMO

Technologies for peripheral nerve stimulation have conventionally relied on the anatomic placement of electrodes adjacent to subsets of sensory fibres or motor fibres that selectively target an end effector. Here, we demonstrate the use of optogenetics to directly target the innervating fibres of an end effector by relying on retrograde transfection of adeno-associated virus serotype 6 to restrict axonal opsin expression to the desired fibre targets. By using an in vivo screen in rats, we identify the first channelrhodopsins as well as a halorhodopsin that respond to red light in the peripheral nerve. Combining two channelrhodopsins with spectrally distinct activation profiles allowed us to drive opposing muscle activity via two-colour illumination of the same mixed nerve. We also show halorhodopsin-mediated reductions in electrically evoked muscle tremor spectrally optimized for deep peripheral nerves. Our non-invasive peripheral neurostimulator with targeted multi-fascicle resolution enables scientific and clinical exploration, such as motor control in paralysis, biomimetic sensation feedback for amputees and targeted inhibition of muscle tremor.


Assuntos
Channelrhodopsins/metabolismo , Optogenética , Nervos Periféricos/metabolismo , Animais , Axônios/metabolismo , Channelrhodopsins/genética , Cor , Dependovirus/genética , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Membro Posterior/patologia , Luz , Opsinas/genética , Opsinas/metabolismo , Nervos Periféricos/efeitos da radiação , Ratos , Ratos Endogâmicos F344 , Estimulação Elétrica Nervosa Transcutânea
4.
PLoS One ; 12(7): e0181264, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28708877

RESUMO

The median raphe region (MRR) is believed to control the fear circuitry indirectly, by influencing the encoding and retrieval of fear memories by amygdala, hippocampus and prefrontal cortex. Here we show that in addition to this established role, MRR stimulation may alone elicit the emergence of remote but not recent fear memories. We substituted electric shocks with optic stimulation of MRR in C57BL/6N male mice in an optogenetic conditioning paradigm and found that stimulations produced agitation, but not fear, during the conditioning trial. Contextual fear, reflected by freezing was not present the next day, but appeared after a 7 days incubation. The optogenetic silencing of MRR during electric shocks ameliorated conditioned fear also seven, but not one day after conditioning. The optogenetic stimulation patterns (50Hz theta burst and 20Hz) used in our tests elicited serotonin release in vitro and lead to activation primarily in the periaqueductal gray examined by c-Fos immunohistochemistry. Earlier studies demonstrated that fear can be induced acutely by stimulation of several subcortical centers, which, however, do not generate persistent fear memories. Here we show that the MRR also elicits fear, but this develops slowly over time, likely by plastic changes induced by the area and its connections. These findings assign a specific role to the MRR in fear learning. Particularly, we suggest that this area is responsible for the durable sensitization of fear circuits towards aversive contexts, and by this, it contributes to the persistence of fear memories. This suggests the existence a bottom-up control of fear circuits by the MRR, which complements the top-down control exerted by the medial prefrontal cortex.


Assuntos
Encéfalo/fisiologia , Animais , Comportamento Animal , Eletrochoque , Medo/fisiologia , Halorrodopsinas/metabolismo , Imuno-Histoquímica , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Substância Cinzenta Periaquedutal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Serotonina/metabolismo , Gravação em Vídeo
5.
J Gene Med ; 18(1-3): 27-37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26824337

RESUMO

BACKGROUND: The ability to manipulate the activity of interneurons with optogenetic tools offers the possibility of interfering with diseases caused by altered neuronal inhibition and synchrony, including epilepsy and schizophrenia. To develop vectors for therapeutic approaches, targeting optogenetic constructs to interneurons is therefore a key requirement. We investigated whether the interneuron-specific promoters glutamic acid decarboxylase (GAD)67 and cholecystokinin (CCK) allowed targeted lentiviral delivery of opsins to interneurons as a whole, or specifically CCK+ interneurons. METHODS: We generated lentiviral (LV) plasmids encoding channelrhodopsin (ChR2) and halorhodopsin (NpHR) tagged with fluorophores and driven by GAD67 or CCK promoters. Adeno-associated virus (AAV) and LV vectors carrying opsins driven by pyramidal cell promoters were used as controls. We transduced neuronal cultures and rodent brain in vivo, immunostained specimens 6-8 weeks after in vivo injection and 7-14 days after in vitro transduction, and evaluated volume and specificity of expression by confocal microscopy. RESULTS: In vitro, 90% (19/21) of LV-CCK-NpHR2.0-EYFP expressing neurons were CCK+. In vivo, LV-GAD67-ChR2-mCherry was expressed in 2.6% (5/193), LV-GAD67-NpHR2.0-EYFP in approximately 15% (43/279) and LV-CCK-NpHR2.0-EYFP in 47% (9/19) of hippocampal GABA+ interneurons. GAD67 vectors expressed in larger volumes than CCK-driven constructs. AAV vector controls achieved the largest expression volumes. CONCLUSIONS: LV-CCK-NpHR2.0-EYFP may be useful for targeting CCK+ interneurons in culture. GAD67/CCK-driven lentiviral constructs are expressed in vivo, although expression is not specific for interneurons. Overall, expression levels are low compared to opsins driven by pyramidal cell promoters. A better understanding of GAD67 and CCK promoter structure or alternative techniques is required to reliably target opsins to interneurons using viral vectors.


Assuntos
Colecistocinina/genética , Glutamato Descarboxilase/genética , Hipocampo/metabolismo , Interneurônios/metabolismo , Transdução Genética , Animais , Células Cultivadas , Channelrhodopsins , Dependovirus , Vetores Genéticos , Halorrodopsinas/metabolismo , Humanos , Lentivirus , Masculino , Optogenética , Regiões Promotoras Genéticas , Células Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Proc Natl Acad Sci U S A ; 111(42): 15238-43, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25246561

RESUMO

Left-right asymmetries have likely evolved to make optimal use of bilaterian nervous systems; however, little is known about the synaptic and circuit mechanisms that support divergence of function between equivalent structures in each hemisphere. Here we examined whether lateralized hippocampal memory processing is present in mice, where hemispheric asymmetry at the CA3-CA1 pyramidal neuron synapse has recently been demonstrated, with different spine morphology, glutamate receptor content, and synaptic plasticity, depending on whether afferents originate in the left or right CA3. To address this question, we used optogenetics to acutely silence CA3 pyramidal neurons in either the left or right dorsal hippocampus while mice performed hippocampus-dependent memory tasks. We found that unilateral silencing of either the left or right CA3 was sufficient to impair short-term memory. However, a striking asymmetry emerged in long-term memory, wherein only left CA3 silencing impaired performance on an associative spatial long-term memory task, whereas right CA3 silencing had no effect. To explore whether synaptic properties intrinsic to the hippocampus might contribute to this left-right behavioral asymmetry, we investigated the expression of hippocampal long-term potentiation. Following the induction of long-term potentiation by high-frequency electrical stimulation, synapses between CA3 and CA1 pyramidal neurons were strengthened only when presynaptic input originated in the left CA3, confirming an asymmetry in synaptic properties. The dissociation of hippocampal long-term memory function between hemispheres suggests that memory is routed via distinct left-right pathways within the mouse hippocampus, and provides a promising approach to help elucidate the synaptic basis of long-term memory.


Assuntos
Região CA3 Hipocampal/fisiologia , Memória/fisiologia , Animais , Comportamento Animal , Mapeamento Encefálico , Dependovirus , Inativação Gênica , Halorrodopsinas/metabolismo , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/fisiologia , Células Piramidais/fisiologia , Memória Espacial , Sinapses/fisiologia
7.
PLoS One ; 8(4): e62013, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23637949

RESUMO

Epilepsy is a devastating disease, currently treated with medications, surgery or electrical stimulation. None of these approaches is totally effective and our ability to control seizures remains limited and complicated by frequent side effects. The emerging revolutionary technique of optogenetics enables manipulation of the activity of specific neuronal populations in vivo with exquisite spatiotemporal resolution using light. We used optogenetic approaches to test the role of hippocampal excitatory neurons in the lithium-pilocarpine model of acute elicited seizures in awake behaving rats. Hippocampal pyramidal neurons were transduced in vivo with a virus carrying an enhanced halorhodopsin (eNpHR), a yellow light activated chloride pump, and acute seizure progression was then monitored behaviorally and electrophysiologically in the presence and absence of illumination delivered via an optical fiber. Inhibition of those neurons with illumination prior to seizure onset significantly delayed electrographic and behavioral initiation of status epilepticus, and altered the dynamics of ictal activity development. These results reveal an essential role of hippocampal excitatory neurons in this model of ictogenesis and illustrate the power of optogenetic approaches for elucidation of seizure mechanisms. This early success in controlling seizures also suggests future therapeutic avenues.


Assuntos
Optogenética/métodos , Estado Epiléptico/terapia , Animais , Modelos Animais de Doenças , Eletroencefalografia , Expressão Gênica , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Hipocampo/metabolismo , Masculino , Optogenética/efeitos adversos , Células Piramidais/metabolismo , Ratos , Convulsões/genética , Convulsões/fisiopatologia , Convulsões/terapia , Estado Epiléptico/genética , Estado Epiléptico/fisiopatologia , Transdução Genética
8.
Biol Chem ; 394(2): 271-80, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23134970

RESUMO

Channelrhodopsin-2 is a light-gated cation channel from the green alga Chlamydomonas reinhardtii. It is functional in animal cells and therefore widely used for light-activated depolarization, especially in neurons. To achieve a fully functional protein, the chromophore all-trans-retinal is needed. It has not been investigated whether or not the apoprotein is stable without its cofactor until now. Here we show that channelopsin-2 (Chop2, protein without bound retinal) is much more prone to degradation than channelrhodopsin-2 (protein with retinal). Constructs of Chop2 fused to yellow fluorescent protein (Chop2::YFP) in the absence and presence of retinal confirm this observation by exhibiting strongly differing fluorescence. We present mutants of Chop2 with highly increased stability in the absence of retinal. Substitution of threonine 159 with aromatic amino acids causes enhanced resistance to degradation in the absence of retinal, which is confirmed by fluorescence intensity, the increase in photocurrents on the addition of retinal to previously expressed protein, and Western blot analysis. Exchanging threonine 159 with cysteine, however, increases photocurrents due to better binding of retinal, without obvious stabilization against degradation of the retinal-free opsin. We also show that the light-activated hyperpolarizing chloride pump halorhodopsin from Natronomonas pharaonis (NpHR) is not prone to retinal-dependent degradation.


Assuntos
Proteínas de Transporte/metabolismo , Mutação , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Resistência a Medicamentos , Halorrodopsinas/metabolismo , Halorrodopsinas/efeitos da radiação , Luz , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oócitos/efeitos da radiação , Retinaldeído/farmacologia , Xenopus laevis
9.
PLoS One ; 6(4): e18452, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21483674

RESUMO

The superficial layer of the superior colliculus (sSC) receives visual inputs via two different pathways: from the retina and the primary visual cortex. However, the functional significance of each input for the operation of the sSC circuit remains to be identified. As a first step toward understanding the functional role of each of these inputs, we developed an optogenetic method to specifically suppress the synaptic transmission in the retino-tectal pathway. We introduced enhanced halorhodopsin (eNpHR), a yellow light-sensitive, membrane-targeting chloride pump, into mouse retinal ganglion cells (RGCs) by intravitreously injecting an adeno-associated virus serotype-2 vector carrying the CMV-eNpHR-EYFP construct. Several weeks after the injection, whole-cell recordings made from sSC neurons in slice preparations revealed that yellow laser illumination of the eNpHR-expressing retino-tectal axons, putatively synapsing onto the recorded cells, effectively inhibited EPSCs evoked by electrical stimulation of the optic nerve layer. We also showed that sSC spike activities elicited by visual stimulation were significantly reduced by laser illumination of the sSC in anesthetized mice. These results indicate that photo-activation of eNpHR expressed in RGC axons enables selective blockade of retino-tectal synaptic transmission. The method established here can most likely be applied to a variety of brain regions for studying the function of individual inputs to these regions.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Halorrodopsinas/metabolismo , Luz , Transmissão Sináptica/efeitos da radiação , Córtex Visual/fisiologia , Córtex Visual/efeitos da radiação , Animais , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Axônios/metabolismo , Axônios/efeitos da radiação , Expressão Gênica , Células HEK293 , Halobacteriaceae , Halorrodopsinas/genética , Humanos , Lasers , Camundongos , Camundongos Endogâmicos C57BL , Estimulação Luminosa , Células Ganglionares da Retina/citologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/efeitos da radiação , Colículos Superiores/citologia , Colículos Superiores/metabolismo , Colículos Superiores/fisiologia , Colículos Superiores/efeitos da radiação , Córtex Visual/citologia , Córtex Visual/metabolismo
11.
Science ; 329(5990): 413-7, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20576849

RESUMO

Retinitis pigmentosa refers to a diverse group of hereditary diseases that lead to incurable blindness, affecting two million people worldwide. As a common pathology, rod photoreceptors die early, whereas light-insensitive, morphologically altered cone photoreceptors persist longer. It is unknown if these cones are accessible for therapeutic intervention. Here, we show that expression of archaebacterial halorhodopsin in light-insensitive cones can substitute for the native phototransduction cascade and restore light sensitivity in mouse models of retinitis pigmentosa. Resensitized photoreceptors activate all retinal cone pathways, drive sophisticated retinal circuit functions (including directional selectivity), activate cortical circuits, and mediate visually guided behaviors. Using human ex vivo retinas, we show that halorhodopsin can reactivate light-insensitive human photoreceptors. Finally, we identified blind patients with persisting, light-insensitive cones for potential halorhodopsin-based therapy.


Assuntos
Terapia Genética , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia , Retinose Pigmentar/terapia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Potenciais Evocados Visuais , Vetores Genéticos , Halobacteriaceae/genética , Humanos , Luz , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Retina/fisiologia , Células Ganglionares da Retina/fisiologia , Retinose Pigmentar/fisiopatologia , Técnicas de Cultura de Tecidos , Transfecção , Visão Ocular , Vias Visuais/fisiologia
12.
BMC Evol Biol ; 7: 79, 2007 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-17511874

RESUMO

BACKGROUND: The type 1 (microbial) rhodopsins are a diverse group of photochemically reactive proteins that display a broad yet patchy distribution among the three domains of life. Recent work indicates that this pattern is likely the result of lateral gene transfer (LGT) of rhodopsin genes between major lineages, and even across domain boundaries. Within the lineage in which the microbial rhodopsins were initially discovered, the haloarchaea, a similar patchy distribution is observed. In this initial study, we assess the roles of LGT and gene loss in the evolution of haloarchaeal rhodopsin ion pump genes, using phylogenetics and comparative genomics approaches. RESULTS: Mapping presence/absence of rhodopsins onto the phylogeny of the RNA polymerase B' subunit (RpoB') of the haloarchaea supports previous notions that rhodopsins are patchily distributed. The phylogeny for the bacteriorhodopsin (BR) protein revealed two discrepancies in comparison to the RpoB' marker, while the halorhodopsin (HR) tree showed incongruence to both markers. Comparative analyses of bacteriorhodopsin-linked regions of five haloarchaeal genomes supported relationships observed in the BR tree, and also identified two open reading frames (ORFs) that were more frequently linked to the bacteriorhodopsin gene than those genes previously shown to be important to the function and expression of BR. CONCLUSION: The evidence presented here reveals a complex evolutionary history for the haloarchaeal rhodopsins, with both LGT and gene loss contributing to the patchy distribution of rhodopsins within this group. Similarities between the BR and RpoB' phylogenies provide supportive evidence for the presence of bacteriorhodopsin in the last common ancestor of haloarchaea. Furthermore, two loci that we have designated bacterio-opsin associated chaperone (bac) and bacterio-opsin associated protein (bap) are inferred to have important roles in BR biogenesis based on frequent linkage and co-transfer with bacteriorhodopsin genes.


Assuntos
RNA Polimerases Dirigidas por DNA/genética , Evolução Molecular , Halobacteriaceae/genética , Bombas de Íon/genética , Rodopsinas Microbianas/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Bacteriorodopsinas/genética , Bacteriorodopsinas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Transferência Genética Horizontal , Genes Arqueais , Genoma Arqueal , Halobacteriaceae/metabolismo , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Bombas de Íon/metabolismo , Filogenia , Rodopsinas Microbianas/metabolismo , Proteína com Valosina
13.
Biochemistry ; 46(18): 5349-57, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17432830

RESUMO

A complex of photoreceptor phoborhodopsin (ppR; also called sensory rhodopsin II) and its cognate halobacterial transducer II (pHtrII) existing in the plasma membrane mediates the light signal to the cytoplasm in the earliest step of negative phototaxis in Natronomonas pharaonis. We have investigated the dynamics of the light-induced conformational changes of the ppR/pHtrII(1-159) complex formed in the presence of 0.1% n-dodecyl beta-d-maltoside (DDM) by a fluorescence resonance energy transfer (FRET) based method. Fluorescence donor and acceptor dyes were linked to cysteine residues genetically introduced at given positions in pHtrII and ppR. The light-induced FRET efficiency changes for various pairs of dye-labeled cysteine residues were determined to examine dynamics of movements of given residues in the transmembrane and the linker region including the HAMP domain in pHtrII induced by photoexcitation of ppR. Upon flash excitation of ppR, FRET efficiency changed depending on pairs of the labeled cysteine residues. The distances between V185 in ppR and the five given residues (102 through 141) in the pHtrII linker region estimated from the FRET efficiency increased by 0.3-0.8 A; on the other hand, the distances between S31 in ppR and the five residues in pHtrII decreased. The changes arose within 70 ms (the dead time of instrument) and decayed at a rate of 1.1 +/- 0.2 s. Azide significantly increased the decay rate of light-induced FRET efficiency changes by accelerating the decay of the M state of ppR. The decay rate of FRET efficiency changes coincided with the rate of recovery of the ppR to the initial state but not the decay of the M state. We conclude that the light-induced conformational change of pHtrII occurs before, at the formation or during the M state, and its relaxation is coupled tightly with the decay of the O state of ppR in the 1:1 complex formed in the DDM micelle.


Assuntos
Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Glucosídeos/metabolismo , Halorrodopsinas/química , Halorrodopsinas/metabolismo , Luz , Rodopsinas Sensoriais/química , Rodopsinas Sensoriais/metabolismo , Termodinâmica , Visão Ocular , Detergentes , Transferência Ressonante de Energia de Fluorescência , Glucosídeos/química , Micelas , Fotólise , Conformação Proteica
14.
J Biol Chem ; 281(45): 34239-45, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16968701

RESUMO

Sensory rhodopsin II (SRII), a receptor for negative phototaxis in haloarchaea, transmits light signals through changes in protein-protein interaction with its transducer HtrII. Light-induced structural changes throughout the SRII-HtrII interface, which spans the periplasmic region, membrane-embedded domains, and cytoplasmic domains near the membrane, have been identified by several studies. Here we demonstrate by site-specific mutagenesis and analysis of phototaxis behavior that two residues in SRII near the membrane-embedded interface (Tyr174 on helix F and Thr204 on helix G) are essential for signaling by the SRII-HtrII complex. These residues, which are the first in SRII shown to be required for phototaxis function, provide biological significance to the previous observation that the hydrogen bond between them is strengthened upon the formation of the earliest SRII photointermediate (SRII(K)) only when SRII is complexed with HtrII. Here we report frequency changes of the S-H stretch of a cysteine substituted for SRII Thr204 in the signaling state intermediates of the SRII photocycle, as well as an influence of HtrII on the hydrogen bond strength, supporting a direct role of the hydrogen bond in SRII-HtrII signal relay chemistry. Our results suggest that the light signal is transmitted to HtrII from the energized interhelical hydrogen bond between Thr204 and Tyr174, which is located at both the retinal chromophore pocket and in helices F and G that form the membrane-embedded interaction surface to the signal-bearing second transmembrane helix of HtrII. The results argue for a critical process in signal relay occurring at this membrane interfacial region of the complex.


Assuntos
Halorrodopsinas/química , Rodopsinas Sensoriais/química , Transdução de Sinais , Treonina/química , Tirosina/química , Membrana Celular , Halobacterium salinarum/química , Halobacterium salinarum/genética , Halorrodopsinas/genética , Halorrodopsinas/metabolismo , Ligação de Hidrogênio , Mutagênese Sítio-Dirigida , Plasmídeos , Rodopsinas Sensoriais/genética , Rodopsinas Sensoriais/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Treonina/genética , Tirosina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA