Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 958
Filtrar
1.
Sci Rep ; 14(1): 21028, 2024 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251630

RESUMEN

Novel stimulation methods are needed to overcome the limitations of contemporary cochlear implants. Optogenetics is a technique that confers light sensitivity to neurons via the genetic introduction of light-sensitive ion channels. By controlling neural activity with light, auditory neurons can be activated with higher spatial precision. Understanding the behaviour of opsins at high stimulation rates is an important step towards their translation. To elucidate this, we compared the temporal characteristics of auditory nerve and inferior colliculus responses to optogenetic, electrical, and combined optogenetic-electrical stimulation in virally transduced mice expressing one of two channelrhodopsins, ChR2-H134R or ChIEF, at stimulation rates up to 400 pulses per second (pps). At 100 pps, optogenetic responses in ChIEF mice demonstrated higher fidelity, less change in latency, and greater response stability compared to responses in ChR2-H134R mice, but not at higher rates. Combined stimulation improved the response characteristics in both cohorts at 400 pps, although there was no consistent facilitation of electrical responses. Despite these results, day-long stimulation (up to 13 h) led to severe and non-recoverable deterioration of the optogenetic responses. The results of this study have significant implications for the translation of optogenetic-only and combined stimulation techniques for hearing loss.


Asunto(s)
Vías Auditivas , Channelrhodopsins , Estimulación Eléctrica , Optogenética , Animales , Optogenética/métodos , Ratones , Vías Auditivas/fisiología , Vías Auditivas/metabolismo , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Estimulación Eléctrica/métodos , Colículos Inferiores/fisiología , Colículos Inferiores/metabolismo , Nervio Coclear/fisiología , Nervio Coclear/metabolismo , Cinética , Implantes Cocleares
2.
Mol Cell ; 84(18): 3530-3544.e6, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39232582

RESUMEN

Channelrhodopsins are microbial light-gated ion channels that can control the firing of neurons in response to light. Among several cation channelrhodopsins identified in Guillardia theta (GtCCRs), GtCCR4 has higher light sensitivity than typical channelrhodopsins. Furthermore, GtCCR4 shows superior properties as an optogenetic tool, such as minimal desensitization. Our structural analyses of GtCCR2 and GtCCR4 revealed that GtCCR4 has an outwardly bent transmembrane helix, resembling the conformation of activated G-protein-coupled receptors. Spectroscopic and electrophysiological comparisons suggested that this helix bend in GtCCR4 omits channel recovery time and contributes to high light sensitivity. An electrophysiological comparison of GtCCR4 and the well-characterized optogenetic tool ChRmine demonstrated that GtCCR4 has superior current continuity and action-potential spike generation with less invasiveness in neurons. We also identified highly active mutants of GtCCR4. These results shed light on the diverse structures and dynamics of microbial rhodopsins and demonstrate the strong optogenetic potential of GtCCR4.


Asunto(s)
Bacteriorodopsinas , Luz , Neuronas , Optogenética , Optogenética/métodos , Animales , Neuronas/metabolismo , Neuronas/efectos de la radiación , Bacteriorodopsinas/metabolismo , Bacteriorodopsinas/genética , Bacteriorodopsinas/química , Humanos , Células HEK293 , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Channelrhodopsins/química , Potenciales de Acción , Criptófitas/genética , Criptófitas/metabolismo , Mutación , Activación del Canal Iónico/efectos de la radiación , Relación Estructura-Actividad
3.
Stroke ; 55(10): 2502-2509, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39234742

RESUMEN

BACKGROUND: Functional activation of the focal ischemic brain has been reported to improve outcomes by augmenting collateral blood flow. However, functional activation also increases metabolic demand and might thereby worsen outcomes. Indeed, preclinical and clinical reports have been conflicting. Here, we tested the effect of functional activation during acute ischemic stroke using distal middle cerebral artery occlusion in anesthetized mice. METHODS: Using transgenic mice expressing channelrhodopsin-2 in neurons, we delivered functional activation using physiological levels of transcranial optogenetic stimulation of the moderately ischemic cortex (ie, penumbra), identified using real-time full-field laser speckle perfusion imaging during a 1-hour distal microvascular clip of the middle cerebral artery. Neuronal activation was confirmed using evoked field potentials, and infarct volumes were measured in tissue slices 48 hours later. RESULTS: Optogenetic stimulation of the penumbra was associated with more than 2-fold larger infarcts than stimulation of the contralateral homotopic region and the sham stimulation group (n=10, 7, and 9; 11.0±5.6 versus 5.1±4.3 versus 4.1±3.7 mm3; P=0.008, 1-way ANOVA). Identical stimulation in wild-type mice that do not express channelrhodopsin-2 did not have an effect. Optogenetic stimulation was associated with a small increase in penumbral perfusion that did not explain enlarged infarcts. CONCLUSIONS: Our data suggest that increased neuronal activity during acute focal arterial occlusions can be detrimental, presumably due to increased metabolic demand, and may have implications for the clinical management of hyperacute stroke patients.


Asunto(s)
Accidente Cerebrovascular Isquémico , Ratones Transgénicos , Optogenética , Animales , Ratones , Accidente Cerebrovascular Isquémico/fisiopatología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Isquemia Encefálica/fisiopatología , Neuronas/metabolismo , Circulación Cerebrovascular/fisiología , Ratones Endogámicos C57BL
4.
PLoS One ; 19(9): e0309553, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39241014

RESUMEN

Cation conducting channelrhodopsins (ChRs) are a popular tool used in optogenetics to control the activity of excitable cells and tissues using light. ChRs with altered ion selectivity are in high demand for use in different cell types and for other specialized applications. However, a detailed mechanism of ion permeation in ChRs is not fully resolved. Here, we use complementary experimental and computational methods to uncover the mechanisms of cation transport and valence selectivity through the channelrhodopsin chimera, C1C2, in the high- and low-conducting open states. Electrophysiology measurements identified a single-residue substitution within the central gate, N297D, that increased Ca2+ permeability vs. Na+ by nearly two-fold at peak current, but less so at stationary current. We then developed molecular models of dimeric wild-type C1C2 and N297D mutant channels in both open states and calculated the PMF profiles for Na+ and Ca2+ permeation through each protein using well-tempered/multiple-walker metadynamics. Results of these studies agree well with experimental measurements and demonstrate that the pore entrance on the extracellular side differs from original predictions and is actually located in a gap between helices I and II. Cation transport occurs via a relay mechanism where cations are passed between flexible carboxylate sidechains lining the full length of the pore by sidechain swinging, like a monkey swinging on vines. In the mutant channel, residue D297 enhances Ca2+ permeability by mediating the handoff between the central and cytosolic binding sites via direct coordination and sidechain swinging. We also found that altered cation binding affinities at both the extracellular entrance and central binding sites underly the distinct transport properties of the low-conducting open state. This work significantly advances our understanding of ion selectivity and permeation in cation channelrhodopsins and provides the insights needed for successful development of new ion-selective optogenetic tools.


Asunto(s)
Calcio , Channelrhodopsins , Simulación de Dinámica Molecular , Sodio , Sodio/metabolismo , Calcio/metabolismo , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Channelrhodopsins/química , Animales , Transporte Iónico , Humanos , Células HEK293 , Activación del Canal Iónico
5.
Biochemistry (Mosc) ; 89(8): 1392-1401, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39245452

RESUMEN

Optogenetics, the method of light-controlled regulation of cellular processes is based on the use of the channelrhodopsins that directly generate photoinduced currents. Most of the channelrhodopsin genes have been identified in the green microalgae Chlorophyta, and the demand for increasing the number of functionally characterized channelrhodopsins and the diversity of their photochemical parameters keeps growing. We performed the expression analysis of cation channelrhodopsin (CCR) genes in natural isolates of microalgae of the genera Haematococcus and Bracteacoccus from the unique Arctic Circle region. The identified full-length CCR transcript of H. lacustris is the product of alternative splicing and encodes the Hl98CCR2 protein with no photochemical activity. The 5'-partial fragment of the B. aggregatus CCR transcript encodes the Ba34CCR protein containing a conserved TM1-TM7 membrane domain and a short cytosolic fragment. Upon heterologous expression of the TM1-TM7 fragment in CHO-K1 cell culture, light-dependent current generation was observed with the parameters corresponding to those of the CCR. The first discovered functional channelrhodopsin of Bracteacoccus has no close CCR homologues and may be of interest as a candidate for optogenetics.


Asunto(s)
Channelrhodopsins , Chlorophyta , Chlorophyta/genética , Chlorophyta/metabolismo , Animales , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Células CHO , Cricetulus , Optogenética/métodos , Luz
6.
J Phys Chem B ; 128(36): 8613-8627, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39207723

RESUMEN

Channelrhodopsins are light-gated ion channels with a retinal chromophore found in microbes and are widely used in optogenetics, a field of neuroscience that utilizes light to regulate neuronal activity. GtACR1, an anion conducting channelrhodopsin derived from Guillardia theta, has attracted attention for its application as a neuronal silencer in optogenetics because of its high conductivity and selectivity. However, atomistic mechanisms of channel photoactivation and ion conduction have not yet been elucidated. In the present study, we investigated the molecular characteristics of GtACR1 and its photoactivation processes by molecular simulations. The QM/MM RWFE-SCF method which combines highly accurate quantum chemistry calculations with long-time molecular dynamics (MD) simulations were used to model protein structures of the wild-type and mutants with different protonation states of key groups and to calculate absorption energies for verification of the models. The QM/MM modeling together with MD simulations of free-energy calculations favors protonation of a key counterion carboxyl group of Asp234 with a strong binding of a chloride ion in the extracellular pocket in the dark state. A channel open state was also successfully modeled by the QM/MM RWFE-SCF free-energy optimizations, providing atomistic insights into the channel activation mechanism.


Asunto(s)
Simulación de Dinámica Molecular , Protones , Teoría Cuántica , Channelrhodopsins/química , Channelrhodopsins/metabolismo , Aniones/química , Aniones/metabolismo , Procesos Fotoquímicos
7.
Nature ; 633(8031): 872-877, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39198644

RESUMEN

Early plant responses to different stress situations often encompass cytosolic Ca2+ increases, plasma membrane depolarization and the generation of reactive oxygen species1-3. However, the mechanisms by which these signalling elements are translated into defined physiological outcomes are poorly understood. Here, to study the basis for encoding of specificity in plant signal processing, we used light-gated ion channels (channelrhodopsins). We developed a genetically engineered channelrhodopsin variant called XXM 2.0 with high Ca2+ conductance that enabled triggering cytosolic Ca2+ elevations in planta. Plant responses to light-induced Ca2+ influx through XXM 2.0 were studied side by side with effects caused by an anion efflux through the light-gated anion channelrhodopsin ACR1 2.04. Although both tools triggered membrane depolarizations, their activation led to distinct plant stress responses: XXM 2.0-induced Ca2+ signals stimulated production of reactive oxygen species and defence mechanisms; ACR1 2.0-mediated anion efflux triggered drought stress responses. Our findings imply that discrete Ca2+ signals and anion efflux serve as triggers for specific metabolic and transcriptional reprogramming enabling plants to adapt to particular stress situations. Our optogenetics approach unveiled that within plant leaves, distinct physiological responses are triggered by specific ion fluxes, which are accompanied by similar electrical signals.


Asunto(s)
Arabidopsis , Calcio , Channelrhodopsins , Luz , Optogenética , Especies Reactivas de Oxígeno , Especies Reactivas de Oxígeno/metabolismo , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Calcio/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Sequías , Estrés Fisiológico/genética , Citosol/metabolismo , Aniones/metabolismo , Señalización del Calcio
8.
Nat Commun ; 15(1): 7292, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39181878

RESUMEN

Channelrhodopsins are popular optogenetic tools in neuroscience, but remain poorly understood mechanistically. Here we report the cryo-EM structures of channelrhodopsin-2 (ChR2) from Chlamydomonas reinhardtii and H. catenoides kalium channelrhodopsin (KCR1). We show that ChR2 recruits an endogenous N-retinylidene-PE-like molecule to a previously unidentified lateral retinal binding pocket, exhibiting a reduced light response in HEK293 cells. In contrast, H. catenoides kalium channelrhodopsin (KCR1) binds an endogenous retinal in its canonical retinal binding pocket under identical condition. However, exogenous ATR reduces the photocurrent magnitude of wild type KCR1 and also inhibits its leaky mutant C110T. Our results uncover diverse retinal chromophores with distinct binding patterns for channelrhodopsins in mammalian cells, which may further inspire next generation optogenetics for complex tasks such as cell fate control.


Asunto(s)
Channelrhodopsins , Chlamydomonas reinhardtii , Optogenética , Células HEK293 , Humanos , Chlamydomonas reinhardtii/metabolismo , Chlamydomonas reinhardtii/genética , Optogenética/métodos , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Channelrhodopsins/química , Microscopía por Crioelectrón , Retinaldehído/metabolismo , Retinaldehído/química , Unión Proteica , Sitios de Unión , Rodopsina/metabolismo , Rodopsina/química , Rodopsina/genética , Luz
9.
J Zhejiang Univ Sci B ; 25(8): 656-671, 2024 Aug 15.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-39155779

RESUMEN

Optogenetics combines optics and genetic engineering to control specific gene expression and biological functions and has the advantages of precise spatiotemporal control, noninvasiveness, and high efficiency. Genetically modified photosensory sensors are engineered into proteins to modulate conformational changes with light stimulation. Therefore, optogenetic techniques can provide new insights into oral biological processes at different levels, ranging from the subcellular and cellular levels to neural circuits and behavioral models. Here, we introduce the origins of optogenetics and highlight the recent progress of optogenetic approaches in oral and craniofacial research, focusing on the ability to apply optogenetics to the study of basic scientific neural mechanisms and to establish different oral behavioral test models in vivo (orofacial movement, licking, eating, and drinking), such as channelrhodopsin (ChR), archaerhodopsin (Arch), and halorhodopsin from Natronomonas pharaonis (NpHR). We also review the synergic and antagonistic effects of optogenetics in preclinical studies of trigeminal neuralgia and maxillofacial cellulitis. In addition, optogenetic tools have been used to control the neurogenic differentiation of dental pulp stem cells in translational studies. Although the scope of optogenetic tools is increasing, there are limited large animal experiments and clinical studies in dental research. Potential future directions include exploring therapeutic strategies for addressing loss of taste in patients with coronavirus disease 2019 (COVID-19), studying oral bacterial biofilms, enhancing craniomaxillofacial and periodontal tissue regeneration, and elucidating the possible pathogenesis of dry sockets, xerostomia, and burning mouth syndrome.


Asunto(s)
Optogenética , Optogenética/métodos , Humanos , Animales , Channelrhodopsins/genética , Channelrhodopsins/metabolismo
10.
Brain Nerve ; 76(7): 835-842, 2024 Jul.
Artículo en Japonés | MEDLINE | ID: mdl-38970320

RESUMEN

All-optical methods that provide deeper understanding of neural activity are currently being developed. Optogenetics is a biological technique useful to control neuronal activity or life phenomena using light. Microbial rhodopsins are light-activated membrane proteins used as optogenetic tools. Microbial rhodopsins such as channelrhodopsin2 (ChR2) consist of seven-pass transmembrane proteins with a covalently bound retinal. Light absorption is followed by photoisomerization of the all-trans retinal to a 13-cis configuration and subsequent conformational changes in the molecule, with consequent permeability of the channel structure to ions. Recent studies have reported the discovery of microbial rhodopsins with novel functions. Microbial rhodopsin diversity has also increased. We describe the characteristics of microbial rhodopsins used as optogenetic tools and the latest research in this domain.


Asunto(s)
Optogenética , Optogenética/métodos , Humanos , Animales , Luz , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Rodopsinas Microbianas/metabolismo , Rodopsina/metabolismo , Rodopsina/genética
11.
Am J Physiol Cell Physiol ; 327(3): C716-C727, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39010839

RESUMEN

Gap junctions are channels that allow for direct transmission of electrical signals between cells. However, the ability of one cell to be impacted or controlled by other cells through gap junctions remains unclear. In this study, heterocellular coupling between ON α retinal ganglion cells (α-RGCs) and displaced amacrine cells (ACs) in the mouse retina was used as a model. The impact of the extent of coupling of interconnected ACs on the synchronized firing between coupled ON α-RGC-AC pair was investigated using the dopamine 1 receptor (D1R) antagonist-SCH23390 and agonist-SKF38393. It was observed that the synchronized firing between the ON α-RGC-ACs pairs was increased by the D1R antagonist SCH23390, whereas it was eradicated by the agonist SKF38393. Subsequently, the signaling drive was investigated by infecting coupled ON α-RGC-AC pairs with the channelrhodopsin-2(ChR2) mutation L132C engineered to enhance light sensitivities. The results demonstrated that the spikes of ON α-RGCs (without ChR2) could be triggered by ACs (with ChR2) through the gap junction, and vice versa. Furthermore, it was observed that ON α-RGCs stimulated with 3-10 Hz currents by whole cell patch could elicit synchronous spikes in the coupled ACs, and vice versa. This provided direct evidence that the firing of one cell could be influenced by another cell through gap junctions. However, this phenomenon was not observed between OFF α-RGC pairs. The study implied that the synchronized firing between ON α-RGC-AC pairs could potentially be affected by the coupling of interconnected ACs. Additionally, one cell type could selectively control the firing of another cell type, thereby forcefully transmitting information. The key role of gap junctions in synchronizing firing and driving cells between α-RGCs and coupled ACs in the mouse retina was highlighted.NEW & NOTEWORTHY This study investigates the role of gap junctions in transmitting electrical signals between cells and their potential for cell control. Using ON α retinal ganglion cells (α-RGCs) and amacrine cells (ACs) in the mouse retina, the researchers find that the extent of coupling between ACs affects synchronized firing. Bidirectional signaling occurs between ACs and ON α-RGCs through gap junctions.


Asunto(s)
Potenciales de Acción , Células Amacrinas , Uniones Comunicantes , Células Ganglionares de la Retina , Animales , Células Amacrinas/metabolismo , Células Amacrinas/fisiología , Células Amacrinas/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/fisiología , Ratones , Uniones Comunicantes/metabolismo , Uniones Comunicantes/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Ratones Endogámicos C57BL , Retina/metabolismo , Retina/fisiología , 2,3,4,5-Tetrahidro-7,8-dihidroxi-1-fenil-1H-3-benzazepina/farmacología , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/agonistas , Comunicación Celular/fisiología , Masculino , Channelrhodopsins/metabolismo , Channelrhodopsins/genética
12.
Biochim Biophys Acta Bioenerg ; 1865(4): 149148, 2024 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-38906314

RESUMEN

Channelrhodopsins (CRs) are used as key tools in optogenetics, and novel CRs, either found from nature or engineered by mutation, have greatly contributed to the development of optogenetics. Recently CRs were discovered from viruses, and crystal structure of a viral CR, OLPVR1, reported a very similar water-containing hydrogen-bonding network near the retinal Schiff base to that of a light-driven proton-pump bacteriorhodopsin (BR). In both OLPVR1 and BR, nearly planar pentagonal cluster structures are comprised of five oxygen atoms, three oxygens from water molecules and two oxygens from the Schiff base counterions. The planar pentagonal cluster stabilizes a quadrupole, two positive charges at the Schiff base and an arginine, and two negative charges at the counterions, and thus plays important roles in light-gated channel function of OLPVR1 and light-driven proton pump function of BR. Despite similar pentagonal cluster structures, present FTIR analysis revealed different hydrogen-bonding networks between OLPVR1 and BR. The hydrogen bond between the protonated Schiff base and a water is stronger in OLPVR1 than in BR, and internal water molecules donate hydrogen bonds much weaker in OLPVR1 than in BR. In OLPVR1, the bridged water molecule between the Schiff base and counterions forms hydrogen bonds to D76 and D200 equally, while the hydrogen-bonding interaction is much stronger to D85 than to D212 in BR. The present interpretation is supported by the mutation results, where D76 and D200 equally work as the Schiff base counterions in OLPVR1, but D85 is the primary counterion in BR. This work reports highly sensitive hydrogen-bonding network in the Schiff base region, which would be closely related to each function through light-induced alterations of the network.


Asunto(s)
Enlace de Hidrógeno , Channelrhodopsins/química , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Espectroscopía Infrarroja por Transformada de Fourier , Bases de Schiff/química , Bacteriorodopsinas/química , Bacteriorodopsinas/metabolismo , Bacteriorodopsinas/genética , Agua/química , Agua/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/genética , Modelos Moleculares
13.
Neuroscience ; 551: 276-289, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38838978

RESUMEN

Transient nigrostriatal dopaminergic signalling is well known for its role in reinforcement learning and increasingly so for its role in the initiation of voluntary movement. However, how transient bursts of dopamine modulate voluntary movement remains unclear, likely due to the heterogeneity of the nigrostriatal system, the focus of optogenetic studies on locomotion at sub-sec time intervals, and the overlapping roles of phasic dopamine in behaviour and novelty signalling. In this study we investigated how phasic activity in the lateral substantia nigra pars compacta (lateral SNc) over time affects voluntary behaviours during exploration. Using a transgenic mouse model of both sexes expressing channelrhodopsin (ChR2) in dopamine transporter-expressing cells, we stimulated the lateral SNc while mice explored an open field over two consecutive days. We found that phasic activation of the lateral SNc induced an increase in exploratory behaviours including horizontal movement activity, locomotion initiation, and rearing specifically on the first open field exposure, but not on the second day. In addition, stimulated animals did not habituate to the same extent as their ChR2-negative counterparts, as indicated by a lack of decrease in baseline activity. These findings suggest that rather than prompting voluntary movement in general, phasic nigrostriatal dopamine prompts context-appropriate behaviours. In addition, dopamine signalling that modulates movement acts over longer timescales than the transient signal, affecting behaviour even after the signal has ended.


Asunto(s)
Neuronas Dopaminérgicas , Conducta Exploratoria , Habituación Psicofisiológica , Ratones Transgénicos , Sustancia Negra , Animales , Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/metabolismo , Conducta Exploratoria/fisiología , Masculino , Sustancia Negra/fisiología , Sustancia Negra/metabolismo , Femenino , Habituación Psicofisiológica/fisiología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Ratones , Optogenética , Locomoción/fisiología , Ratones Endogámicos C57BL , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Actividad Motora/fisiología
14.
J Neurophysiol ; 131(6): 1226-1239, 2024 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-38691531

RESUMEN

Mitral/tufted cells (M/TCs) form complex local circuits with interneurons in the olfactory bulb and are powerfully inhibited by these interneurons. The horizontal limb of the diagonal band of Broca (HDB), the only GABAergic/inhibitory source of centrifugal circuit with the olfactory bulb, is known to target olfactory bulb interneurons, and we have shown targeting also to olfactory bulb glutamatergic neurons in vitro. However, the net efficacy of these circuits under different patterns of activation in vivo and the relative balance between the various targeted intact local and centrifugal circuits was the focus of this study. Here channelrhodopsin-2 (ChR2) was expressed in HDB GABAergic neurons to investigate the short-term plasticity of HDB-activated disinhibitory rebound excitation of M/TCs. Optical activation of HDB interneurons increased spontaneous M/TC firing without odor presentation and increased odor-evoked M/TC firing. HDB activation induced disinhibitory rebound excitation (burst or cluster of spiking) in all classes of M/TCs. This excitation was frequency dependent, with short-term facilitation only at higher HDB stimulation frequency (5 Hz and above). However, frequency-dependent HDB regulation was more potent in the deeper layer M/TCs compared with more superficial layer M/TCs. In all neural circuits the balance between inhibition and excitation in local and centrifugal circuits plays a critical functional role, and this patterned input-dependent regulation of inhibitory centrifugal inputs to the olfactory bulb may help maintain the precise balance across the populations of output neurons in different environmental odors, putatively to sharpen the enhancement of tuning specificity of individual or classes of M/TCs to odors.NEW & NOTEWORTHY Neuronal local circuits in the olfactory bulb are modulated by centrifugal long circuits. In vivo study here shows that inhibitory horizontal limb of the diagonal band of Broca (HDB) modulates all five types of mitral/tufted cells (M/TCs), by direct inhibitory circuits HDB → M/TCs and indirect disinhibitory long circuits HDB → interneurons → M/TCs. The HDB net effect exerts excitation in all types of M/TCs but more powerful in deeper layer output neurons as HDB activation frequency increases, which may sharpen the tuning specificity of classes of M/TCs to odors during sensory processing.


Asunto(s)
Interneuronas , Bulbo Olfatorio , Bulbo Olfatorio/fisiología , Bulbo Olfatorio/citología , Animales , Interneuronas/fisiología , Ratones , Neuronas GABAérgicas/fisiología , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Masculino , Ratones Endogámicos C57BL , Potenciales de Acción/fisiología , Inhibición Neural/fisiología , Femenino , Optogenética
15.
J Phys Chem Lett ; 15(21): 5788-5794, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38780133

RESUMEN

Channelrhodopsin (ChR) and heliorhodopsin (HeR) are microbial rhodopsins with similar structures but different circular dichroism (CD) spectra: ChR shows biphasic negative and positive bands, whereas HeR shows a single positive band. We explored the physicochemical factors underlying these differences through computational methods. Using the exciton model based on first-principles computations, we obtained the CD spectra of ChR and HeR. The obtained spectra indicate that the protein dimer structures and the quantum mechanical treatment of the retinal chromophore and its interacting amino acids are crucial for accurately reproducing the experimental spectra. Further calculations revealed that the sign of the excitonic coupling was opposite between the ChR and HeR dimers, which was attributed to the contrasting second term of the orientation factor between the two retinal chromophores. These findings demonstrate that slight variations in the intermolecular orientation of the two chromophores can result in significant differences in the CD spectral shape.


Asunto(s)
Dicroismo Circular , Channelrhodopsins/química , Multimerización de Proteína , Teoría Cuántica , Modelos Moleculares
16.
Biophys J ; 123(12): 1735-1750, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762755

RESUMEN

The light-gated anion channelrhodopsin GtACR1 is an important optogenetic tool for neuronal silencing. Its photochemistry, including its photointermediates, is poorly understood. The current mechanistic view presumes BR-like kinetics and assigns the open channel to a blue-absorbing L intermediate. Based on time-resolved absorption and electrophysiological data, we recently proposed a red-absorbing spectral form for the open channel state. Here, we report the results of a comprehensive kinetic analysis of the spectroscopic data combined with channel current information. The time evolutions of the spectral forms derived from the spectroscopic data are inconsistent with the single chain mechanism and are analyzed within the concept of parallel photocycles. The spectral forms partitioned into conductive and nonconductive parallel cycles are assigned to intermediate states. Rejecting reversible connections between conductive and nonconductive channel states leads to kinetic schemes with two independent conductive states corresponding to the fast- and slow-decaying current components. The conductive cycle is discussed in terms of a single cycle and two parallel cycles. The reaction mechanisms and reaction rates for the wild-type protein, the A75E, and the low-conductance D234N and S97E protein variants are derived. The parallel cycles of channelrhodopsin kinetics, its relation to BR photocycle, and the role of the M intermediate in channel closure are discussed.


Asunto(s)
Activación del Canal Iónico , Cinética , Rodopsina/metabolismo , Rodopsina/química , Rodopsina/genética , Animales , Aniones/metabolismo , Luz , Modelos Biológicos , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Channelrhodopsins/química
17.
Hear Res ; 447: 109009, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38670009

RESUMEN

We recently reported that the central nucleus of the inferior colliculus (the auditory midbrain) is innervated by glutamatergic pyramidal cells originating not only in auditory cortex (AC), but also in multiple 'non-auditory' regions of the cerebral cortex. Here, in anaesthetised rats, we used optogenetics and electrical stimulation, combined with recording in the inferior colliculus to determine the functional influence of these descending connections. Specifically, we determined the extent of monosynaptic excitation and the influence of these descending connections on spontaneous activity in the inferior colliculus. A retrograde virus encoding both green fluorescent protein (GFP) and channelrhodopsin (ChR2) injected into the central nucleus of the inferior colliculus (ICc) resulted in GFP expression in discrete groups of cells in multiple areas of the cerebral cortex. Light stimulation of AC and primary motor cortex (M1) caused local activation of cortical neurones and increased the firing rate of neurones in ICc indicating a direct excitatory input from AC and M1 to ICc with a restricted distribution. In naïve animals, electrical stimulation at multiple different sites within M1, secondary motor, somatosensory, and prefrontal cortices increased firing rate in ICc. However, it was notable that stimulation at some adjacent sites failed to influence firing at the recording site in ICc. Responses in ICc comprised singular spikes of constant shape and size which occurred with a short, and fixed latency (∼ 5 ms) consistent with monosynaptic excitation of individual ICc units. Increasing the stimulus current decreased the latency of these spikes, suggesting more rapid depolarization of cortical neurones, and increased the number of (usually adjacent) channels on which a monosynaptic spike was seen, suggesting recruitment of increasing numbers of cortical neurons. Electrical stimulation of cortical regions also evoked longer latency, longer duration increases in firing activity, comprising multiple units with spikes occurring with significant temporal jitter, consistent with polysynaptic excitation. Increasing the stimulus current increased the number of spikes in these polysynaptic responses and increased the number of channels on which the responses were observed, although the magnitude of the responses always diminished away from the most activated channels. Together our findings indicate descending connections from motor, somatosensory and executive cortical regions directly activate small numbers of ICc neurones and that this in turn leads to extensive polysynaptic activation of local circuits within the ICc.


Asunto(s)
Corteza Auditiva , Vías Auditivas , Estimulación Eléctrica , Colículos Inferiores , Corteza Motora , Optogenética , Corteza Somatosensorial , Sinapsis , Animales , Colículos Inferiores/fisiología , Corteza Somatosensorial/fisiología , Corteza Auditiva/fisiología , Corteza Motora/fisiología , Vías Auditivas/fisiología , Sinapsis/fisiología , Masculino , Neuronas/fisiología , Ratas Sprague-Dawley , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Femenino , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Ratas
18.
Curr Gene Ther ; 24(3): 208-216, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676313

RESUMEN

Hearing loss is a prevalent sensory impairment significantly affecting communication and quality of life. Traditional approaches for hearing restoration, such as cochlear implants, have limitations in frequency resolution and spatial selectivity. Optogenetics, an emerging field utilizing light-sensitive proteins, offers a promising avenue for addressing these limitations and revolutionizing hearing rehabilitation. This review explores the methods of introducing Channelrhodopsin- 2 (ChR2), a key light-sensitive protein, into cochlear cells to enable optogenetic stimulation. Viral- mediated gene delivery is a widely employed technique in optogenetics. Selecting a suitable viral vector, such as adeno-associated viruses (AAV), is crucial in efficient gene delivery to cochlear cells. The ChR2 gene is inserted into the viral vector through molecular cloning techniques, and the resulting viral vector is introduced into cochlear cells via direct injection or round window membrane delivery. This allows for the expression of ChR2 and subsequent light sensitivity in targeted cells. Alternatively, direct cell transfection offers a non-viral approach for ChR2 delivery. The ChR2 gene is cloned into a plasmid vector, which is then combined with transfection agents like liposomes or nanoparticles. This mixture is applied to cochlear cells, facilitating the entry of the plasmid DNA into the target cells and enabling ChR2 expression. Optogenetic stimulation using ChR2 allows for precise and selective activation of specific neurons in response to light, potentially overcoming the limitations of current auditory prostheses. Moreover, optogenetics has broader implications in understanding the neural circuits involved in auditory processing and behavior. The combination of optogenetics and gene delivery techniques provides a promising avenue for improving hearing restoration strategies, offering the potential for enhanced frequency resolution, spatial selectivity, and improved auditory perception.


Asunto(s)
Percepción Auditiva , Terapia Genética , Vectores Genéticos , Pérdida Auditiva , Optogenética , Optogenética/métodos , Humanos , Terapia Genética/métodos , Percepción Auditiva/genética , Vectores Genéticos/genética , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Channelrhodopsins/genética , Dependovirus/genética , Técnicas de Transferencia de Gen , Animales , Implantes Cocleares
19.
Nat Commun ; 15(1): 3480, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658537

RESUMEN

The analysis of neural circuits has been revolutionized by optogenetic methods. Light-gated chloride-conducting anion channelrhodopsins (ACRs)-recently emerged as powerful neuron inhibitors. For cells or sub-neuronal compartments with high intracellular chloride concentrations, however, a chloride conductance can have instead an activating effect. The recently discovered light-gated, potassium-conducting, kalium channelrhodopsins (KCRs) might serve as an alternative in these situations, with potentially broad application. As yet, KCRs have not been shown to confer potent inhibitory effects in small genetically tractable animals. Here, we evaluated the utility of KCRs to suppress behavior and inhibit neural activity in Drosophila, Caenorhabditis elegans, and zebrafish. In direct comparisons with ACR1, a KCR1 variant with enhanced plasma-membrane trafficking displayed comparable potency, but with improved properties that include reduced toxicity and superior efficacy in putative high-chloride cells. This comparative analysis of behavioral inhibition between chloride- and potassium-selective silencing tools establishes KCRs as next-generation optogenetic inhibitors for in vivo circuit analysis in behaving animals.


Asunto(s)
Caenorhabditis elegans , Neuronas , Optogenética , Pez Cebra , Animales , Caenorhabditis elegans/genética , Neuronas/metabolismo , Neuronas/fisiología , Optogenética/métodos , Channelrhodopsins/metabolismo , Channelrhodopsins/genética , Humanos , Drosophila , Canales de Potasio/metabolismo , Canales de Potasio/genética , Cloruros/metabolismo , Animales Modificados Genéticamente , Conducta Animal , Células HEK293 , Drosophila melanogaster
20.
Nat Commun ; 15(1): 3525, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664445

RESUMEN

Soft bioelectronic devices exhibit motion-adaptive properties for neural interfaces to investigate complex neural circuits. Here, we develop a fabrication approach through the control of metamorphic polymers' amorphous-crystalline transition to miniaturize and integrate multiple components into hydrogel bioelectronics. We attain an about 80% diameter reduction in chemically cross-linked polyvinyl alcohol hydrogel fibers in a fully hydrated state. This strategy allows regulation of hydrogel properties, including refractive index (1.37-1.40 at 480 nm), light transmission (>96%), stretchability (139-169%), bending stiffness (4.6 ± 1.4 N/m), and elastic modulus (2.8-9.3 MPa). To exploit the applications, we apply step-index hydrogel optical probes in the mouse ventral tegmental area, coupled with fiber photometry recordings and social behavioral assays. Additionally, we fabricate carbon nanotubes-PVA hydrogel microelectrodes by incorporating conductive nanomaterials in hydrogel for spontaneous neural activities recording. We enable simultaneous optogenetic stimulation and electrophysiological recordings of light-triggered neural activities in Channelrhodopsin-2 transgenic mice.


Asunto(s)
Hidrogeles , Ratones Transgénicos , Optogenética , Polímeros , Alcohol Polivinílico , Animales , Alcohol Polivinílico/química , Ratones , Hidrogeles/química , Optogenética/métodos , Polímeros/química , Nanotubos de Carbono/química , Área Tegmental Ventral/fisiología , Microelectrodos , Masculino , Channelrhodopsins/metabolismo , Channelrhodopsins/química , Channelrhodopsins/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...