Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Science ; 381(6665): 1480-1487, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37769108

RESUMEN

After heart injury, dead heart muscle is replaced by scar tissue. Fibroblasts can electrically couple with myocytes, and changes in fibroblast membrane potential can lead to myocyte excitability, which suggests that fibroblast-myocyte coupling in scar tissue may be responsible for arrhythmogenesis. However, the physiologic relevance of electrical coupling of myocytes and fibroblasts and its impact on cardiac excitability in vivo have never been demonstrated. We genetically engineered a mouse that expresses the optogenetic cationic channel ChR2 (H134R) exclusively in cardiac fibroblasts. After myocardial infarction, optical stimulation of scar tissue elicited organ-wide cardiac excitation and induced arrhythmias in these animals. Complementing computational modeling with experimental approaches, we showed that gap junctional and ephaptic coupling, in a synergistic yet functionally redundant manner, excited myocytes coupled to fibroblasts.


Asunto(s)
Arritmias Cardíacas , Channelrhodopsins , Cicatriz , Fibroblastos , Miocitos Cardíacos , Animales , Ratones , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Cicatriz/patología , Cicatriz/fisiopatología , Fibroblastos/fisiología , Miocitos Cardíacos/fisiología , Channelrhodopsins/genética , Channelrhodopsins/fisiología , Optogenética , Conexina 43/genética , Conexina 43/fisiología , Técnicas de Inactivación de Genes
2.
Commun Biol ; 4(1): 578, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990694

RESUMEN

Channelrhodopsins are widely used in optogenetic applications. High photocurrents and low current inactivation levels are desirable. Two parallel photocycles evoked by different retinal conformations cause cation-conducting channelrhodopsin-2 (CrChR2) inactivation: one with efficient conductivity; one with low conductivity. Given the longer half-life of the low conducting photocycle intermediates, which accumulate under continuous illumination, resulting in a largely reduced photocurrent. Here, we demonstrate that for channelrhodopsin-1 of the cryptophyte Guillardia theta (GtACR1), the highly conducting C = N-anti-photocycle was the sole operating cycle using time-resolved step-scan FTIR spectroscopy. The correlation between our spectroscopic measurements and previously reported electrophysiological data provides insights into molecular gating mechanisms and their role in the characteristic high photocurrents. The mechanistic importance of the central constriction site amino acid Glu-68 is also shown. We propose that canceling out the poorly conducting photocycle avoids the inactivation observed in CrChR2, and anticipate that this discovery will advance the development of optimized optogenetic tools.


Asunto(s)
Aniones/química , Channelrhodopsins/fisiología , Criptófitas/fisiología , Fenómenos Electrofisiológicos , Activación del Canal Iónico , Luz , Optogenética , Espectrofotometría
3.
Methods Mol Biol ; 2191: 201-220, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32865747

RESUMEN

Zebrafish are an excellent model organism to study many aspects of vertebrate sensory encoding and behavior. Their escape responses begin with a C-shaped body bend followed by several swimming bouts away from the potentially threatening stimulus. This highly stereotyped motor behavior provides a model for studying startle reflexes and the neural circuitry underlying multisensory encoding and locomotion. Channelrhodopsin (ChR2) can be expressed in the lateral line and ear hair cells of zebrafish and can be excited in vivo to elicit these rapid forms of escape. Here we review our methods for studying transgenic ChR2-expressing zebrafish larvae, including screening for positive expression of ChR2 and recording field potentials and high-speed videos of optically evoked escape responses. We also highlight important features of the acquired data and provide a brief review of other zebrafish research that utilizes or has the potential to benefit from ChR2 and optogenetics.


Asunto(s)
Channelrhodopsins/genética , Potenciales Evocados/genética , Neuronas/metabolismo , Optogenética/métodos , Animales , Animales Modificados Genéticamente/genética , Channelrhodopsins/fisiología , Potenciales Evocados/fisiología , Células Ciliadas Auditivas/metabolismo , Larva/fisiología , Locomoción/genética , Locomoción/fisiología , Neuronas/patología , Reflejo de Sobresalto/fisiología , Natación/fisiología , Pez Cebra/genética , Pez Cebra/fisiología
4.
Invest Ophthalmol Vis Sci ; 61(10): 44, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32841313

RESUMEN

Purpose: Disruption in blood supply to active retinal circuits is the earliest hallmark of diabetic retinopathy (DR) and has been primarily attributed to vascular deficiency. However, accumulating evidence supports an early role for a disrupted neuronal function in blood flow impairment. Here, we tested the hypothesis that selectively stimulating cholinergic neurons could restore neurovascular signaling to preserve the capillary circulation in DR. Methods: We used wild type (wt) and choline acetyltransferase promoter (ChAT)-channelrhodopsin-2 (ChR2) mice expressing ChR2 exclusively in cholinergic cells. Mice were made diabetic by streptozotocin (STZ) injections. Two to 3 months after the last STZ injection, the rate of capillary blood flow was measured in vivo within each retinal vascular layer using high speed two-photon imaging. Measurements were done at baseline and following ChR2-driven activation of retinal cholinergic interneurons, the sole source of the vasodilating neurotransmitter acetylcholine. After recordings, retinas were collected and assessed for physiological and structural features. Results: In retinal explants from ChAT-ChR2 mice, we found that channelrhodopsin2 was selectively expressed in all cholinergic amacrine cells. Its direct activation by blue light led to dilation of adjacent retinal capillaries. In living diabetic ChAT-ChR2 animals, basal capillary blood flow was significantly higher than in diabetic mice without channelrhodopsin. However, optogenetic stimulation with blue light did not result in flickering light-induced functional hyperemia, suggesting a necessity for a concerted neurovascular interaction. Conclusions: These findings provide direct support to the utility and efficacy of an optogenetic approach for targeting selective retinal circuits to treat DR and its complications.


Asunto(s)
Células Amacrinas/fisiología , Neuronas Colinérgicas/fisiología , Retinopatía Diabética/terapia , Optogenética/métodos , Células Amacrinas/patología , Animales , Channelrhodopsins/metabolismo , Channelrhodopsins/fisiología , Neuronas Colinérgicas/patología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Flujo Sanguíneo Regional , Retina/patología , Vasos Retinianos/patología , Vasos Retinianos/fisiología
5.
Proc Natl Acad Sci U S A ; 117(34): 20920-20925, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32788371

RESUMEN

In plants, environmental stressors trigger plasma membrane depolarizations. Being electrically interconnected via plasmodesmata, proper functional dissection of electrical signaling by electrophysiology is basically impossible. The green alga Chlamydomonas reinhardtii evolved blue light-excited channelrhodopsins (ChR1, 2) to navigate. When expressed in excitable nerve and muscle cells, ChRs can be used to control the membrane potential via illumination. In Arabidopsis plants, we used the algal ChR2-light switches as tools to stimulate plasmodesmata-interconnected photosynthetic cell networks by blue light and monitor the subsequent plasma membrane electrical responses. Blue-dependent stimulations of ChR2 expressing mesophyll cells, resting around -160 to -180 mV, reproducibly depolarized the membrane potential by 95 mV on average. Following excitation, mesophyll cells recovered their prestimulus potential not without transiently passing a hyperpolarization state. By combining optogenetics with voltage-sensing microelectrodes, we demonstrate that plant plasma membrane AHA-type H+-ATPase governs the gross repolarization process. AHA2 protein biochemistry and functional expression analysis in Xenopus oocytes indicates that the capacity of this H+ pump to recharge the membrane potential is rooted in its voltage- and pH-dependent functional anatomy. Thus, ChR2 optogenetics appears well suited to noninvasively expose plant cells to signal specific depolarization signatures. From the responses we learn about the molecular processes, plants employ to channel stress-associated membrane excitations into physiological responses.


Asunto(s)
Membrana Celular/metabolismo , Channelrhodopsins/metabolismo , Bombas de Protones/metabolismo , Adenosina Trifosfatasas/metabolismo , Proteínas Algáceas/metabolismo , Channelrhodopsins/fisiología , Chlamydomonas reinhardtii/metabolismo , Color , Concentración de Iones de Hidrógeno , Luz , Potenciales de la Membrana/fisiología , Optogenética/métodos , Bombas de Protones/fisiología , Rodopsina/metabolismo , Transducción de Señal
6.
Nat Commun ; 11(1): 2716, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32483118

RESUMEN

How do brain mechanisms create maladaptive attractions? Here intense maladaptive attractions are created in laboratory rats by pairing optogenetic channelrhodopsin (ChR2) stimulation of central nucleus of amygdala (CeA) in rats with encountering either sucrose, cocaine, or a painful shock-delivering object. We find that pairings make the respective rats pursue either sucrose exclusively, or cocaine exclusively, or repeatedly self-inflict shocks. CeA-induced maladaptive attractions, even to the painful shock-rod, recruit mesocorticolimbic incentive-related circuitry. Shock-associated cues also gain positive incentive value and are pursued. Yet the motivational effects of paired CeA stimulation can be reversed to negative valence in a Pavlovian fear learning situation, where CeA ChR2 pairing increases defensive reactions. Finally, CeA ChR2 valence can be switched to neutral by pairing with innocuous stimuli. These results reveal valence plasticity and multiple modes for motivation via mesocorticolimbic circuitry under the control of CeA activation.


Asunto(s)
Encéfalo/fisiología , Núcleo Amigdalino Central/fisiología , Channelrhodopsins/fisiología , Dolor/fisiopatología , Recompensa , Animales , Núcleo Amigdalino Central/metabolismo , Channelrhodopsins/metabolismo , Cocaína/administración & dosificación , Señales (Psicología) , Femenino , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Motivación/fisiología , Optogenética/métodos , Ratas Sprague-Dawley , Sacarosa/administración & dosificación
7.
Biomed Phys Eng Express ; 6(2): 025003, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33438629

RESUMEN

OBJECTIVE: There is a need for low power, scalable photoelectronic devices and systems for emerging optogenetic needs in neuromodulation. Conventional light emitting diodes (LEDs) are constrained by power and lead-counts necessary for scalability. Organic LEDs (OLEDs) offer an exciting approach to decrease power and lead-counts while achieving high channel counts on thin, flexible substrates that conform to brain surfaces or peripheral neuronal fibers. In this study, we investigate the potential for using OLEDs to modulate neuronal networks cultured in vitro on a transparent microelectrode array (MEA) and subsequently validate neurostimulation in vivo in a transgenic mouse model. APPROACH: Cultured mouse cortical neurons were transfected with light-sensitive opsins such as blue-light sensitive channel-rhodopsin (ChR2) and green-light sensitive chimeric channel-rhodopsin (C1V1tt) and stimulated using blue and green OLEDs (with 455 and 520 nm peak emission spectra respectively) at a power of ~1 mW mm-2 under pulsed conditions. MAIN RESULTS: We demonstrate neuromodulation and optostimulus-locked, single unit-neuronal activity in neurons expressing stimulating opsins (34 units on n = 4 MEAs, each with 16 recordable channels). We also validated the optostimulus-locked response in preliminary experiments in a channel-rhodopsin expressing transgenic mouse model, where at least three isolatable single neuronal cortical units respond to OLED stimulation. SIGNIFICANCE: The above results indicate the feasibility of generating sufficient luminance from OLEDs to perform neuromodulation both in vitro and in vivo. This opens up the possibility of developing thin, flexible OLED films with multiple stimulation sites that can conform to the shape of the neuronal targets in the brain or the peripheral nervous system. However, stability of these OLEDs under chronic conditions still needs to be carefully assessed with appropriate packaging approaches.


Asunto(s)
Channelrhodopsins/fisiología , Electrodos , Luz , Neuronas/fisiología , Optogenética , Estimulación Luminosa/métodos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Neuronas/efectos de la radiación
8.
J Neurosci Res ; 98(3): 471-480, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31544973

RESUMEN

BioLuminescent (BL) light production can modulate neural activity and behavior through co-expressed OptoGenetic (OG) elements, an approach termed "BL-OG." Yet, the relationship between BL-OG effects and bioluminescent photon emission has not been characterized in vivo. Further, the degree to which BL-OG effects strictly depend on optogenetic mechanisms driven by bioluminescent photons is unknown. Crucial to every neuromodulation method is whether the activator shows a dynamic concentration range driving robust, selective, and nontoxic effects. We systematically tested the effects of four key components of the BL-OG mechanism (luciferin, oxidized luciferin, luciferin vehicle, and bioluminescence), and compared these against effects induced by the Luminopsin-3 (LMO3) BL-OG molecule, a fusion of slow burn Gaussia luciferase (sbGLuc) and Volvox ChannelRhodopsin-1 (VChR1). We performed combined bioluminescence imaging and electrophysiological recordings while injecting specific doses of Coelenterazine (substrate for sbGluc), Coelenteramide (CTM, the oxidized product of CTZ), or CTZ vehicle. CTZ robustly drove activity in mice expressing LMO3, with photon production proportional to firing rate. In contrast, low and moderate doses of CTZ, CTM, or vehicle did not modulate activity in mice that did not express LMO3. We also failed to find bioluminescence effects on neural activity in mice expressing an optogenetically nonsensitive LMO3 variant. We observed weak responses to the highest dose of CTZ in control mice, but these effects were significantly smaller than those observed in the LMO3 group. These results show that in neocortex in vivo, there is a large CTZ range wherein BL-OG effects are specific to its active chemogenetic mechanism.


Asunto(s)
Mediciones Luminiscentes , Neocórtex/fisiología , Neuronas/fisiología , Optogenética/métodos , Animales , Channelrhodopsins/fisiología , Femenino , Imidazoles/administración & dosificación , Sustancias Luminiscentes/administración & dosificación , Proteínas Luminiscentes , Masculino , Ratones Endogámicos C57BL , Neocórtex/efectos de los fármacos , Opsinas/fisiología , Pirazinas/administración & dosificación , Reproducibilidad de los Resultados
9.
J Neurosci Res ; 98(3): 422-436, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-30957296

RESUMEN

Although molecular tools for controlling neuronal activity by light have vastly expanded, there are still unmet needs which require development and refinement. For example, light delivery into the brain is still a major practical challenge that hinders potential translation of optogenetics in human patients. In addition, it would be advantageous to manipulate neuronal activity acutely and precisely as well as chronically and non-invasively, using the same genetic construct in animal models. We have previously addressed these challenges by employing bioluminescence and have created a new line of opto-chemogenetic probes termed luminopsins by fusing light-sensing opsins with light-emitting luciferases. In this report, we incorporated Chlamydomonas channelrhodopsin 2 with step-function mutations as the opsin moiety in the new luminopsin fusion protein termed step-function luminopsin (SFLMO). Bioluminescence-induced photocurrent lasted longer than the bioluminescence signal due to very slow deactivation of the mutated channel. In addition, bioluminescence was able to activate most of the channels on the cell surface due to the extremely high light sensitivity of the channel. This efficient channel activation was partly mediated by radiationless bioluminescence resonance energy transfer due to the proximity of luciferase and opsin. To test the utility of SFLMOs in vivo, we transduced the substantia nigra unilaterally via a viral vector in male rats. Injection of the luciferase substrate as well as conventional photostimulation via fiber optics elicited circling behaviors. Thus, SFLMOs expand the current approaches for manipulation of neuronal activity in the brain and add more versatility and practicality to optogenetics in freely behaving animals.


Asunto(s)
Channelrhodopsins , Luciferasas , Neuronas/fisiología , Optogenética/métodos , Animales , Conducta Animal/fisiología , Channelrhodopsins/genética , Channelrhodopsins/fisiología , Femenino , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/fisiología , Proteínas Luminiscentes , Masculino , Potenciales de la Membrana , Cultivo Primario de Células , Ratas Sprague-Dawley , Sustancia Negra/fisiología
10.
J Neurosci Res ; 98(3): 410-421, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-28862809

RESUMEN

Previous work has demonstrated that fusion of a luciferase to an opsin, to create a luminescent opsin or luminopsin, provides a genetically encoded means of manipulating neuronal activity via both chemogenetic and optogenetic approaches. Here we have expanded and refined the versatility of luminopsin tools by fusing an alternative luciferase variant with high light emission, Gaussia luciferase mutant GLucM23, to depolarizing and hyperpolarizing channelrhodopsins with increased light sensitivity. The combination of GLucM23 with Volvox channelrhodopsin-1 produced LMO4, while combining GLucM23 with the anion channelrhodopsin iChloC yielded iLMO4. We found efficient activation of these channelrhodopsins in the presence of the luciferase substrate, as indicated by responses measured in both single neurons and in neuronal populations of mice and rats, as well as by changes in male rat behavior during amphetamine-induced rotations. We conclude that these new luminopsins will be useful for bimodal opto- and chemogenetic analyses of brain function.


Asunto(s)
Channelrhodopsins , Luciferasas , Neuronas/fisiología , Optogenética/métodos , Potenciales de Acción , Adenoviridae/fisiología , Animales , Channelrhodopsins/genética , Channelrhodopsins/fisiología , Femenino , Vectores Genéticos , Células HEK293 , Hipocampo/fisiología , Humanos , Luciferasas/genética , Luciferasas/fisiología , Masculino , Ratones , Cultivo Primario de Células , Ratas Sprague-Dawley , Volvox/genética
11.
J Neurosci ; 40(1): 159-170, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31694963

RESUMEN

The cerebellum drives motor coordination and sequencing of actions at the millisecond timescale through adaptive control of cerebellar nuclear output. Cerebellar nuclei integrate high-frequency information from both the cerebellar cortex and the two main excitatory inputs of the cerebellum: the mossy fibers and the climbing fiber collaterals. However, how nuclear cells process rate and timing of inputs carried by these inputs is still debated. Here, we investigate the influence of the cerebellar cortical output, the Purkinje cells, on identified cerebellar nuclei neurons in vivo in male mice. Using transgenic mice expressing Channelrhodopsin2 specifically in Purkinje cells and tetrode recordings in the medial nucleus, we identified two main groups of neurons based on the waveform of their action potentials. These two groups of neurons coincide with glutamatergic and GABAergic neurons identified by optotagging after Chrimson expression in VGLUT2-cre and GAD-cre mice, respectively. The glutamatergic-like neurons fire at high rate and respond to both rate and timing of Purkinje cell population inputs, whereas GABAergic-like neurons only respond to the mean population firing rate of Purkinje cells at high frequencies. Moreover, synchronous activation of Purkinje cells can entrain the glutamatergic-like, but not the GABAergic-like, cells over a wide range of frequencies. Our results suggest that the downstream effect of synchronous and rhythmic Purkinje cell discharges depends on the type of cerebellar nuclei neurons targeted.SIGNIFICANCE STATEMENT Motor coordination and skilled movements are driven by the permanent discharge of neurons from the cerebellar nuclei that communicate cerebellar computation to other brain areas. Here, we set out to study how specific subtypes of cerebellar nuclear neurons of the medial nucleus are controlled by Purkinje cells, the sole output of the cerebellar cortex. We could isolate different subtypes of nuclear cell that differentially encode Purkinje cell inhibition. Purkinje cell stimulation entrains glutamatergic projection cells at their firing frequency, whereas GABAergic neurons are only inhibited. These differential coding strategies may favor temporal precision of cerebellar excitatory outputs associated with specific features of movement control while setting the global level of cerebellar activity through inhibition via rate coding mechanisms.


Asunto(s)
Núcleos Cerebelosos/fisiología , Neuronas GABAérgicas/fisiología , Ácido Glutámico/fisiología , Células de Purkinje/fisiología , Potenciales de Acción , Vías Aferentes/fisiología , Anestesia , Animales , Núcleos Cerebelosos/citología , Channelrhodopsins/fisiología , Genes Reporteros , Glutamato Descarboxilasa/genética , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Destreza Motora , Neuronas/fisiología , Optogenética , Factores de Tiempo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Vigilia
12.
Nat Methods ; 16(11): 1176-1184, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31611694

RESUMEN

We engineered light-gated channelrhodopsins (ChRs) whose current strength and light sensitivity enable minimally invasive neuronal circuit interrogation. Current ChR tools applied to the mammalian brain require intracranial surgery for transgene delivery and implantation of fiber-optic cables to produce light-dependent activation of a small volume of tissue. To facilitate expansive optogenetics without the need for invasive implants, our engineering approach leverages the substantial literature of ChR variants to train statistical models for the design of high-performance ChRs. With Gaussian process models trained on a limited experimental set of 102 functionally characterized ChRs, we designed high-photocurrent ChRs with high light sensitivity. Three of these, ChRger1-3, enable optogenetic activation of the nervous system via systemic transgene delivery. ChRger2 enables light-induced neuronal excitation without fiber-optic implantation; that is, this opsin enables transcranial optogenetics.


Asunto(s)
Channelrhodopsins/genética , Aprendizaje Automático , Optogenética , Ingeniería de Proteínas/métodos , Animales , Channelrhodopsins/fisiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL
13.
J Neurophysiol ; 122(5): 1962-1974, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31533018

RESUMEN

Optogenetics comprise a promising alternative to electrical stimulation for characterization of neural circuits and for the next generation of neural prostheses. Optogenetic stimulation relies on expression of photosensitive microbial proteins in animal cells to initiate a flow of ions into the cells in response to visible light. Here, we generated a novel transgenic mouse model in which we studied the optogenetic activation of spiral ganglion neurons, the primary afferent neurons of the auditory system, and showed a strong optogenetic response, with a similar amplitude as the acoustically evoked response. A twofold increase in the level of channelrhodopsin expression significantly increased the photosensitivity at both the single cell and organismal levels but also partially compromised the native electrophysiological properties of the neurons. The importance of channelrhodopsin expression level to optogenetic stimulation, revealed by these quantitative measurements, will be significant for the characterization of neural circuitry and for the use of optogenetics in neural prostheses.NEW & NOTEWORTHY This study reveals a dose-response relationship between channelrhodopsin expression and optogenetic excitation. Both single cell and organismal responses depend on the expression level of the heterologous protein. Expression level of the opsin is thus an important variable in determining the outcome of an optogenetic experiment. These results are key to the implementation of neural prostheses based on optogenetics, such as next generation cochlear implants, which would use light to elicit a neural response to sound.


Asunto(s)
Channelrhodopsins/fisiología , Cóclea/fisiología , Fenómenos Electrofisiológicos , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Neuronas Aferentes/fisiología , Optogenética , Ganglio Espiral de la Cóclea/fisiología , Animales , Ratones , Ratones Transgénicos , Modelos Animales
14.
J Neurosci ; 39(3): 485-502, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30478035

RESUMEN

It is well known that the posterior parietal cortex (PPC) and frontal motor cortices in primates preferentially control voluntary movements of contralateral limbs. The PPC of rats has been defined based on patterns of thalamic and cortical connectivity. The anatomical characteristics of this area suggest that it may be homologous to the PPC of primates. However, its functional roles in voluntary forelimb movements have not been well understood, particularly in the lateralization of motor limb representation; that is, the limb-specific activity representations for right and left forelimb movements. We examined functional spike activity of the PPC and two motor cortices, the primary motor cortex (M1) and the secondary motor cortex (M2), when head-fixed male rats performed right or left unilateral movements. Unlike primates, PPC neurons in rodents were found to preferentially represent ipsilateral forelimb movements, in contrast to the contralateral preference of M1 and M2 neurons. Consistent with these observations, optogenetic activation of PPC and motor cortices, respectively, evoked ipsilaterally and contralaterally biased forelimb movements. Finally, we examined the effects of optogenetic manipulation on task performance. PPC or M1 inhibition by optogenetic GABA release shifted the behavioral limb preference contralaterally or ipsilaterally, respectively. In addition, weak optogenetic PPC activation, which was insufficient to evoke motor responses by itself, shifted the preference ipsilaterally; although similar M1 activation showed no effects on task performance. These paradoxical observations suggest that the PPC plays evolutionarily different roles in forelimb control between primates and rodents.SIGNIFICANCE STATEMENT In rodents, the primary and secondary motor cortices (M1 and M2, respectively) are involved in voluntary movements with contralateral preference. However, it remains unclear whether and how the posterior parietal cortex (PPC) participates in controlling multiple limb movements. We recorded functional activity from these areas using a behavioral task to monitor movements of the right and left forelimbs separately. PPC neurons preferentially represented ipsilateral forelimb movements and optogenetic PPC activation evoked ipsilaterally biased forelimb movements. Optogenetic PPC inhibition via GABA release shifted the behavioral limb preference contralaterally during task performance, whereas weak optogenetic PPC activation, which was insufficient to evoke motor responses by itself, shifted the preference ipsilaterally. Our findings suggest rodent PPC contributes to ipsilaterally biased motor response and/or planning.


Asunto(s)
Miembro Anterior/fisiología , Lateralidad Funcional/fisiología , Movimiento/fisiología , Lóbulo Parietal/fisiología , Animales , Channelrhodopsins/genética , Channelrhodopsins/fisiología , Condicionamiento Operante , Electromiografía , Masculino , Corteza Motora/fisiología , Optogenética , Técnicas de Placa-Clamp , Desempeño Psicomotor/fisiología , Ratas , Ratas Transgénicas , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/fisiología
15.
eNeuro ; 5(3)2018.
Artículo en Inglés | MEDLINE | ID: mdl-30027111

RESUMEN

Optogenetic inhibition of specific neuronal types in the brain enables analysis of neural circuitry and is promising for the treatment of a number of neurological disorders. Anion channelrhodopsins (ACRs) from the cryptophyte alga Guillardia theta generate larger photocurrents than other available inhibitory optogenetic tools, but more rapid channels are needed for temporally precise inhibition, such as single-spike suppression, of high-frequency firing neurons. Faster ACRs have been reported, but their potential advantages for time-resolved inhibitory optogenetics have not so far been verified in neurons. We report RapACR, nicknamed so for "rapid," an ACR from Rhodomonas salina, that exhibits channel half-closing times below 10 ms and achieves equivalent inhibition at 50-fold lower light intensity in lentivirally transduced cultured mouse hippocampal neurons as the second-generation engineered Cl--conducting channelrhodopsin iC++. The upper limit of the time resolution of neuronal silencing with RapACR determined by measuring the dependence of spiking recovery after photoinhibition on the light intensity was calculated to be 100 Hz, whereas that with the faster of the two G. theta ACRs was 13 Hz. Further acceleration of RapACR channel kinetics was achieved by site-directed mutagenesis of a single residue in transmembrane helix 3 (Thr111 to Cys). We also show that mutation of another ACR (Cys to Ala at the same position) with a greatly extended lifetime of the channel open state acts as a bistable photochromic tool in mammalian neurons. These molecules extend the time domain of optogenetic neuronal silencing while retaining the high light sensitivity of Guillardia ACRs.


Asunto(s)
Channelrhodopsins/fisiología , Activación del Canal Iónico , Neuronas/fisiología , Optogenética/métodos , Potenciales de Acción , Animales , Aniones , Células Cultivadas , Channelrhodopsins/genética , Criptófitas , Células HEK293 , Hipocampo/fisiología , Humanos , Ratones
16.
Biochem Biophys Res Commun ; 496(3): 814-819, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29395082

RESUMEN

Channelrhodopsin-2 (ChR2), a light-activated cation-selective ion channel, has been widely used as a tool in optogenetic research. ChR2 is specifically sensitive to wavelengths less than 550 nm. One of the methods to expand the sensitivity of a channelrhodopsin to a wider range of wavelengths is to express another channelrhodopsin in the cells by the transduction of an additional gene. Here, we report the characteristic features of cells expressing two types of channelrhodopsins, each having different wavelength sensitivities. In HEK293 cells stably expressing ChR2, photocurrents were elicited at stimuli of 400-550 nm, and the wavelength sensitivity range was expanded by the additional transduction of the modified Volvox channelrhodopsin-1 (mVChR1) gene, which has broad wavelength sensitivities, ranging from 400 to 600 nm. However, the photocurrent at 550 nm was lower than that of the mVChR1-expressing cell; moreover, the turning-on and turning-off constants were delayed, and the deactivation rates were decreased. Meanwhile, the response to lower light intensity was improved by the additional gene. Thus, the transduction of an additional gene is a useful method to improve the light and wavelength sensitivities, as well as photocurrent kinetic profiles, of channelrhodopsins.


Asunto(s)
Channelrhodopsins/fisiología , Channelrhodopsins/efectos de la radiación , Activación del Canal Iónico/fisiología , Activación del Canal Iónico/efectos de la radiación , Fototransducción/fisiología , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células HEK293 , Humanos , Cinética , Luz , Dosis de Radiación
17.
Neuron ; 97(2): 434-449.e4, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29307710

RESUMEN

Mesolimbic dopamine (DA) neurons play a central role in motivation and reward processing. Although the activity of these mesolimbic DA neurons is controlled by afferent inputs, little is known about the circuits in which they are embedded. Using retrograde tracing, electrophysiology, optogenetics, and behavioral assays, we identify principles of afferent-specific control in the mesolimbic DA system. Neurons in the medial shell subdivision of the nucleus accumbens (NAc) exert direct inhibitory control over two separate populations of mesolimbic DA neurons by activating different GABA receptor subtypes. In contrast, NAc lateral shell neurons mainly synapse onto ventral tegmental area (VTA) GABA neurons, resulting in disinhibition of DA neurons that project back to the NAc lateral shell. Lastly, we establish a critical role for NAc subregion-specific input to the VTA underlying motivated behavior. Collectively, our results suggest a distinction in the incorporation of inhibitory inputs between different subtypes of mesolimbic DA neurons.


Asunto(s)
Conducta Animal/fisiología , Neuronas Dopaminérgicas/fisiología , Núcleo Accumbens/fisiología , Área Tegmental Ventral/fisiología , Vías Aferentes/fisiología , Animales , Channelrhodopsins/fisiología , Channelrhodopsins/efectos de la radiación , Condicionamiento Operante , Dopamina/fisiología , Neuronas Dopaminérgicas/clasificación , Conducta Exploratoria , Femenino , Neuronas GABAérgicas/fisiología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Motivación , Optogenética , Recompensa , Área Tegmental Ventral/citología
18.
J Neurosci ; 38(6): 1588-1599, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29311142

RESUMEN

Orexin (also known as hypocretin) neurons are considered a key component of the ascending arousal system. They are active during wakefulness, at which time they drive and maintain arousal, and are silent during sleep. Their activity is controlled by long-range inputs from many sources, as well as by more short-range inputs, including from presumptive GABAergic neurons in the lateral hypothalamus/perifornical region (LH/PF). To characterize local GABAergic input to orexin neurons, we used channelrhodopsin-2-assisted circuit mapping in brain slices. We expressed channelrhodopsin-2 in GABAergic neurons (Vgat+) in the LH/PF and recorded from genetically identified surrounding orexin neurons (LH/PFVgat → Orx). We performed all experiments in mice of either sex. Photostimulation of LH/PF GABAergic neurons inhibited the firing of orexin neurons through the release of GABA, evoking GABAA-mediated IPSCs in orexin neurons. These photo-evoked IPSCs were maintained in the presence of TTX, indicating direct connectivity. Carbachol inhibited LH/PFVgat → Orx input through muscarinic receptors. By contrast, application of orexin was without effect on LH/PFVgat → Orx input, whereas dynorphin, another peptide produced by orexin neurons, inhibited LH/PFVgat → Orx input through κ-opioid receptors. Our results demonstrate that orexin neurons are under inhibitory control by local GABAergic neurons and that this input is depressed by cholinergic signaling, unaffected by orexin and inhibited by dynorphin. We propose that local release of dynorphin may, via collaterals, provides a positive feedback to orexin neurons and that, during wakefulness, orexin neurons may be disinhibited by acetylcholine and by their own release of dynorphin.SIGNIFICANCE STATEMENT The lateral hypothalamus contains important wake-promoting cell populations, including orexin-producing neurons. Intermingled with the orexin neurons, there are other cell populations that selectively discharge during nonrapid eye movement or rapid eye movement sleep. Some of these sleep-active neurons release GABA and are thought to inhibit wake-active neurons during rapid eye movement and nonrapid eye movement sleep. However, this hypothesis had not been tested. Here we show that orexin neurons are inhibited by a local GABAergic input. We propose that this local GABAergic input inhibits orexin neurons during sleep but that, during wakefulness, this input is depressed, possibly through cholinergically mediated disinhibition and/or by release of dynorphin from orexin neurons themselves.


Asunto(s)
Neuronas GABAérgicas/fisiología , Área Hipotalámica Lateral/metabolismo , Área Hipotalámica Lateral/fisiología , Orexinas/fisiología , Animales , Mapeo Encefálico , Carbacol/farmacología , Channelrhodopsins/fisiología , Dinorfinas/farmacología , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Área Hipotalámica Lateral/efectos de los fármacos , Masculino , Ratones , Agonistas Muscarínicos/farmacología , Orexinas/farmacología , Sistema Nervioso Parasimpático/fisiología , Estimulación Luminosa , Corteza Prefrontal/metabolismo , Receptores de GABA-A/metabolismo , Tetrodotoxina/farmacología , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/fisiología
19.
Herzschrittmacherther Elektrophysiol ; 29(1): 24-29, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29305704

RESUMEN

Optogenetics is an emerging, interdisciplinary research area which combines genetic and optical technologies to steer and monitor specific biological processes. To this end, light-activated proteins, so-called optogenetic actuators, or fluorescent sensor proteins are genetically targeted to the cells of interest. Light activation can then be used to modulate or record cellular behaviour with high spatiotemporal precision. In cardiac research, optogenetic approaches have been used to unravel heterocellular electrotonic interactions, both in vitro and in situ. Pioneering optogenetic studies with potential relevance for clinical electrophysiology include light-controlled pacing experiments and optical defibrillation studies. However, despite successful implementation in mouse models, clinical applications are not feasible to date; these will require major advances in gene therapy and in optical techniques.


Asunto(s)
Optogenética/métodos , Animales , Estimulación Cardíaca Artificial/métodos , Estimulación Cardíaca Artificial/tendencias , Channelrhodopsins/fisiología , Cardioversión Eléctrica/métodos , Cardioversión Eléctrica/tendencias , Sinapsis Eléctricas/fisiología , Predicción , Humanos , Comunicación Interdisciplinaria , Colaboración Intersectorial , Optogenética/tendencias
20.
J Math Biol ; 76(3): 567-608, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28664220

RESUMEN

We use conductance based neuron models, and the mathematical modeling of optogenetics to define controlled neuron models and we address the minimal time control of these affine systems for the first spike from equilibrium. We apply tools of geometric optimal control theory to study singular extremals, and we implement a direct method to compute optimal controls. When the system is too large to theoretically investigate the existence of singular optimal controls, we observe numerically the optimal bang-bang controls.


Asunto(s)
Potenciales de Acción/fisiología , Channelrhodopsins/fisiología , Modelos Neurológicos , Neuronas/fisiología , Animales , Biología Computacional , Conceptos Matemáticos , Conducción Nerviosa/fisiología , Optogenética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...