Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.840
Filter
1.
Biotechnol J ; 19(5): e2400023, 2024 May.
Article in English | MEDLINE | ID: mdl-38719589

ABSTRACT

The discovery of antibiotics has noticeably promoted the development of human civilization; however, antibiotic resistance in bacteria caused by abusing and overusing greatly challenges human health and food safety. Considering the worsening situation, it is an urgent demand to develop emerging nontraditional technologies or methods to address this issue. With the expanding of synthetic biology, optogenetics exhibits a tempting prospect for precisely regulating gene expression in many fields. Consequently, it is attractive to employ optogenetics to reduce the risk of antibiotic resistance. Here, a blue light-controllable gene expression system was established in Escherichia coli based on a photosensitive DNA-binding protein (EL222). Further, this strategy was successfully applied to repress the expression of ß-lactamase gene (bla) using blue light illumination, resulting a dramatic reduction of ampicillin resistance in engineered E. coli. Moreover, blue light was utilized to induce the expression of the mechanosensitive channel of large conductance (MscL), triumphantly leading to the increase of streptomycin susceptibility in engineered E. coli. Finally, the increased susceptibility of ampicillin and streptomycin was simultaneously induced by blue light in the same E. coli cell, revealing the excellent potential of this strategy in controlling multidrug-resistant (MDR) bacteria. As a proof of concept, our work demonstrates that light can be used as an alternative tool to prolong the use period of common antibiotics without developing new antibiotics. And this novel strategy based on optogenetics shows a promising foreground to combat antibiotic resistance in the future.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Light , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Anti-Bacterial Agents/pharmacology , Optogenetics/methods , Gene Expression Regulation, Bacterial/drug effects , Ampicillin/pharmacology , beta-Lactamases/genetics , beta-Lactamases/metabolism , Drug Resistance, Bacterial/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Streptomycin/pharmacology , Blue Light
2.
Sci Robot ; 9(90): eadi8995, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776378

ABSTRACT

Closed-loop neuroprostheses show promise in restoring motion in individuals with neurological conditions. However, conventional activation strategies based on functional electrical stimulation (FES) fail to accurately modulate muscle force and exhibit rapid fatigue because of their unphysiological recruitment mechanism. Here, we present a closed-loop control framework that leverages physiological force modulation under functional optogenetic stimulation (FOS) to enable high-fidelity muscle control for extended periods of time (>60 minutes) in vivo. We first uncovered the force modulation characteristic of FOS, showing more physiological recruitment and significantly higher modulation ranges (>320%) compared with FES. Second, we developed a neuromuscular model that accurately describes the highly nonlinear dynamics of optogenetically stimulated muscle. Third, on the basis of the optogenetic model, we demonstrated real-time control of muscle force with improved performance and fatigue resistance compared with FES. This work lays the foundation for fatigue-resistant neuroprostheses and optogenetically controlled biohybrid robots with high-fidelity force modulation.


Subject(s)
Muscle Fatigue , Muscle, Skeletal , Optogenetics , Optogenetics/methods , Optogenetics/instrumentation , Animals , Muscle Fatigue/physiology , Muscle, Skeletal/physiology , Humans , Electric Stimulation/instrumentation , Muscle Contraction/physiology , Robotics/instrumentation , Male , Equipment Design , Neural Prostheses , Nonlinear Dynamics
3.
ACS Synth Biol ; 13(5): 1467-1476, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38696739

ABSTRACT

Optogenetics is a powerful tool for spatiotemporal control of gene expression. Several light-inducible gene regulators have been developed to function in bacteria, and these regulatory circuits have been ported to new host strains. Here, we developed and adapted a red-light-inducible transcription factor for Shewanella oneidensis. This regulatory circuit is based on the iLight optogenetic system, which controls gene expression using red light. A thermodynamic model and promoter engineering were used to adapt this system to achieve differential gene expression in light and dark conditions within a S. oneidensis host strain. We further improved the iLight optogenetic system by adding a repressor to invert the genetic circuit and activate gene expression under red light illumination. The inverted iLight genetic circuit was used to control extracellular electron transfer within S. oneidensis. The ability to use both red- and blue-light-induced optogenetic circuits simultaneously was also demonstrated. Our work expands the synthetic biology capabilities in S. oneidensis, which could facilitate future advances in applications with electrogenic bacteria.


Subject(s)
Light , Optogenetics , Promoter Regions, Genetic , Shewanella , Shewanella/genetics , Shewanella/metabolism , Optogenetics/methods , Electron Transport , Promoter Regions, Genetic/genetics , Gene Expression Regulation, Bacterial , Transcription Factors/metabolism , Transcription Factors/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Regulatory Networks/genetics , Synthetic Biology/methods
4.
PLoS Comput Biol ; 20(5): e1012053, 2024 May.
Article in English | MEDLINE | ID: mdl-38709828

ABSTRACT

Monosynaptic connectivity mapping is crucial for building circuit-level models of neural computation. Two-photon optogenetic stimulation, when combined with whole-cell recording, enables large-scale mapping of physiological circuit parameters. In this experimental setup, recorded postsynaptic currents are used to infer the presence and strength of connections. For many cell types, nearby connections are those we expect to be strongest. However, when the postsynaptic cell expresses opsin, optical excitation of nearby cells can induce direct photocurrents in the postsynaptic cell. These photocurrent artifacts contaminate synaptic currents, making it difficult or impossible to probe connectivity for nearby cells. To overcome this problem, we developed a computational tool, Photocurrent Removal with Constraints (PhoRC). Our method is based on a constrained matrix factorization model which leverages the fact that photocurrent kinetics are less variable than those of synaptic currents. We demonstrate on real and simulated data that PhoRC consistently removes photocurrents while preserving synaptic currents, despite variations in photocurrent kinetics across datasets. Our method allows the discovery of synaptic connections which would have been otherwise obscured by photocurrent artifacts, and may thus reveal a more complete picture of synaptic connectivity. PhoRC runs faster than real time and is available as open source software.


Subject(s)
Artifacts , Computational Biology , Models, Neurological , Optogenetics , Optogenetics/methods , Animals , Computational Biology/methods , Synapses/physiology , Mice , Neurons/physiology , Software , Computer Simulation , Algorithms , Patch-Clamp Techniques/methods , Humans
5.
J Diabetes ; 16(6): e13557, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38751366

ABSTRACT

Diabetes mellitus (DM) is a common chronic disease affecting humans globally. It is characterized by abnormally elevated blood glucose levels due to the failure of insulin production or reduction of insulin sensitivity and functionality. Insulin and glucagon-like peptide (GLP)-1 replenishment or improvement of insulin resistance are the two major strategies to treat diabetes. Recently, optogenetics that uses genetically encoded light-sensitive proteins to precisely control cell functions has been regarded as a novel therapeutic strategy for diabetes. Here, we summarize the latest development of optogenetics and its integration with synthetic biology approaches to produce light-responsive cells for insulin/GLP-1 production, amelioration of insulin resistance and neuromodulation of insulin secretion. In addition, we introduce the development of cell encapsulation and delivery methods and smart bioelectronic devices for the in vivo application of optogenetics-based cell therapy in diabetes. The remaining challenges for optogenetics-based cell therapy in the clinical translational study are also discussed.


Subject(s)
Diabetes Mellitus , Optogenetics , Humans , Optogenetics/methods , Diabetes Mellitus/therapy , Animals , Insulin/metabolism , Insulin Resistance , Glucagon-Like Peptide 1 , Cell- and Tissue-Based Therapy/methods , Insulin-Secreting Cells/metabolism
6.
Elife ; 132024 May 15.
Article in English | MEDLINE | ID: mdl-38748470

ABSTRACT

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.


Subject(s)
Cholinergic Neurons , Dopamine , Interneurons , Optogenetics , Dopamine/metabolism , Animals , Interneurons/metabolism , Interneurons/physiology , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Rats , Optogenetics/methods , Motivation , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Acetylcholine/metabolism
7.
Nat Commun ; 15(1): 4306, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773114

ABSTRACT

Heliorhodopsins (HeRs) have been hypothesized to have widespread functions. Recently, the functions for few HeRs have been revealed; however, the hypothetical functions remain largely unknown. Herein, we investigate light-modulation of heterodimeric multidrug resistance ATP-binding cassette transporters (OmrDE) mediated by Omithinimicrobium cerasi HeR. In this study, we classifiy genes flanking the HeR-encoding genes and identify highly conservative residues for protein-protein interactions. Our results reveal that the interaction between OcHeR and OmrDE shows positive cooperatively sequential binding through thermodynamic parameters. Moreover, light-induced OcHeR upregulates OmrDE drug transportation. Hence, the binding may be crucial to drug resistance in O. cerasi as it survives in a drug-containing habitat. Overall, we unveil a function of HeR as regulatory rhodopsin for multidrug resistance. Our findings suggest potential applications in optogenetic technology.


Subject(s)
ATP-Binding Cassette Transporters , Light , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Protein Binding , Rhodopsins, Microbial/metabolism , Rhodopsins, Microbial/genetics , Rhodopsins, Microbial/chemistry , Optogenetics/methods
8.
Sci Rep ; 14(1): 11642, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773346

ABSTRACT

Vision restoration is one of the most promising applications of optogenetics. However, it is limited due to the poor-sensitivity, slow-kinetics and narrow band absorption spectra of opsins. Here, a detailed theoretical study of retinal ganglion neurons (RGNs) expressed with ChRmine, ReaChR, CoChR, CatCh and their mutants, with near monochromatic LEDs, and broadband sunlight, halogen lamp, RGB LED light, and pure white light sources has been presented. All the opsins exhibit improved light sensitivity and larger photocurrent on illuminating with broadband light sources compared to narrow band LEDs. ChRmine allows firing at ambient sunlight (1.5 nW/mm2) and pure white light (1.2 nW/mm2), which is lowest among the opsins considered. The broadband activation spectrum of ChRmine and its mutants is also useful to restore color sensitivity. Although ChRmine exhibits slower turn-off kinetics with broadband light, high-fidelity spikes can be evoked upto 50 Hz. This limit extends upto 80 Hz with the improved hsChRmine mutant although it requires double the irradiance compared to ChRmine. The present study shows that ChRmine and its mutants allow activation of RGNs with ambient light which is useful for goggle-free white light optogenetic retinal prostheses with improved quality of restored vision.


Subject(s)
Light , Optogenetics , Retinal Ganglion Cells , Optogenetics/methods , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/radiation effects , Humans , Mutation , Animals , Opsins/genetics , Opsins/metabolism , Vision, Ocular/physiology
9.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791270

ABSTRACT

At present, there are a variety of different approaches to the targeted regulation of gene expression. However, most approaches are devoted to the activation of gene transcription, and the methods for gene silencing are much fewer in number. In this review, we describe the main systems used for the targeted suppression of gene expression (including RNA interference (RNAi), chimeric transcription factors, chimeric zinc finger proteins, transcription activator-like effectors (TALEs)-based repressors, optogenetic tools, and CRISPR/Cas-based repressors) and their application in eukaryotes-plants and animals. We consider the advantages and disadvantages of each approach, compare their effectiveness, and discuss the peculiarities of their usage in plant and animal organisms. This review will be useful for researchers in the field of gene transcription suppression and will allow them to choose the optimal method for suppressing the expression of the gene of interest depending on the research object.


Subject(s)
CRISPR-Cas Systems , Gene Silencing , Plants , Animals , Plants/genetics , Plants/metabolism , RNA Interference , Transcription Factors/genetics , Transcription Factors/metabolism , Optogenetics/methods , Gene Expression Regulation , Zinc Fingers/genetics
10.
ACS Nano ; 18(17): 11058-11069, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38630984

ABSTRACT

Perioperative neurocognitive disorder (PND) is a common complication in surgical patients. While many interventions to prevent PND have been studied, the availability of treatment methods is limited. Thus, it is crucial to delve into the mechanisms of PND, pinpoint therapeutic targets, and develop effective treatment approaches. In this study, reduced dorsal tenia tecta (DTT) neuronal activity was found to be associated with tibial fracture surgery-induced PND, indicating that a neuronal excitation-inhibition (E-I) imbalance could contribute to PND. Optogenetics in the DTT brain region was conducted using upconversion nanoparticles (UCNPs) with the ability to convert 808 nm near-infrared light to visible wavelengths, which triggered the activation of excitatory neurons with minimal damage in the DTT brain region, thus improving cognitive impairment symptoms in the PND model. Moreover, this noninvasive intervention to modulate E-I imbalance showed a positive influence on mouse behavior in the Morris water maze test, which demonstrates that UCNP-mediated optogenetics is a promising tool for the treatment of neurological imbalance disorders.


Subject(s)
Nanoparticles , Optogenetics , Animals , Optogenetics/methods , Mice , Nanoparticles/chemistry , Male , Maze Learning , Postoperative Cognitive Complications/etiology , Mice, Inbred C57BL , Neurons , Tibial Fractures/surgery , Infrared Rays
11.
Protein Sci ; 33(5): e4993, 2024 May.
Article in English | MEDLINE | ID: mdl-38647395

ABSTRACT

Optogenetic tools (OTs) operating in the far-red and near-infrared (NIR) region offer advantages for light-controlling biological processes in deep tissues and spectral multiplexing with fluorescent probes and OTs acting in the visible range. However, many NIR OTs suffer from background activation in darkness. Through shortening linkers, we engineered a novel NIR OT, iLight2, which exhibits a significantly reduced background activity in darkness, thereby increasing the light-to-dark activation contrast. The resultant optimal configuration of iLight2 components suggests a molecular mechanism of iLight2 action. Using a biliverdin reductase knock-out mouse model, we show that iLight2 exhibits advanced performance in mouse primary cells and deep tissues in vivo. Efficient light-controlled cell migration in wound healing cellular model demonstrates the possibility of using iLight2 in therapy and, overall, positions it as a valuable addition to the NIR OT toolkit for gene transcription applications.


Subject(s)
Optogenetics , Animals , Optogenetics/methods , Mice , Transcription, Genetic , Mice, Knockout , Humans , Infrared Rays
12.
Open Biol ; 14(4): 240001, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38653331

ABSTRACT

Autophagy is a double-edged sword for cells; it can lead to both cell survival and death. Calcium (Ca2+) signalling plays a crucial role in regulating various cellular behaviours, including cell migration, proliferation and death. In this study, we investigated the effects of modulating cytosolic Ca2+ levels on autophagy using chemical and optogenetic methods. Our findings revealed that ionomycin and thapsigargin induce Ca2+ influx to promote autophagy, whereas the Ca2+ chelator BAPTA-AM induces Ca2+ depletion and inhibits autophagy. Furthermore, the optogenetic platform allows the manipulation of illumination parameters, including density, frequency, duty cycle and duration, to create different patterns of Ca2+ oscillations. We used the optogenetic tool Ca2+-translocating channelrhodopsin, which is activated and opened by 470 nm blue light to induce Ca2+ influx. These results demonstrated that high-frequency Ca2+ oscillations induce autophagy. In addition, autophagy induction may involve Ca2+-activated adenosine monophosphate (AMP)-activated protein kinases. In conclusion, high-frequency optogenetic Ca2+ oscillations led to cell death mediated by AMP-activated protein kinase-induced autophagy.


Subject(s)
AMP-Activated Protein Kinases , Autophagy , Calcium , Optogenetics , AMP-Activated Protein Kinases/metabolism , Calcium/metabolism , Calcium Signaling , Enzyme Activation , Ionomycin/pharmacology , Optogenetics/methods , Thapsigargin/pharmacology
13.
Nano Lett ; 24(18): 5403-5412, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38669639

ABSTRACT

The efficacy of electrical stimulation facilitating peripheral nerve regeneration is evidenced extensively, while the associated secondary damage resulting from repeated electrode invasion and indiscriminate stimulation is inevitable. Here, we present an optogenetics strategy that utilizes upconversion nanoparticles (UCNPs) to convert deeply penetrating near-infrared excitation into blue emission, which activates an adeno-associated virus-encoding ChR2 photoresponsive ion channel on cell membranes. The induced Ca2+ flux, similar to the ion flux in the electrical stimulation approach, efficiently regulates viability and proliferation, secretion of nerve growth factor, and neural function of RSC96 cells. Furthermore, deep near-infrared excitation is harnessed to stimulate autologous Schwann cells in situ via a UCNP-composited scaffold, which enhances nerve sprouting and myelination, consequently promoting functional recovery, electrophysiological restoration, and reinnervation of damaged nerves. This developed postoperatively noninvasive optogenetics strategy presents a novel, minimally traumatic, and enduring therapeutic stimulus to effectively promote peripheral nerve repair.


Subject(s)
Nanoparticles , Nerve Regeneration , Optogenetics , Schwann Cells , Sciatic Nerve , Animals , Optogenetics/methods , Nanoparticles/chemistry , Rats , Dependovirus/genetics , Cell Line , Peripheral Nerve Injuries/therapy
14.
ACS Synth Biol ; 13(5): 1424-1433, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38684225

ABSTRACT

The ability to control cellular processes using optogenetics is inducer-limited, with most optogenetic systems responding to blue light. To address this limitation, we leverage an integrated framework combining Lustro, a powerful high-throughput optogenetics platform, and machine learning tools to enable multiplexed control over blue light-sensitive optogenetic systems. Specifically, we identify light induction conditions for sequential activation as well as preferential activation and switching between pairs of light-sensitive split transcription factors in the budding yeast, Saccharomyces cerevisiae. We use the high-throughput data generated from Lustro to build a Bayesian optimization framework that incorporates data-driven learning, uncertainty quantification, and experimental design to enable the prediction of system behavior and the identification of optimal conditions for multiplexed control. This work lays the foundation for designing more advanced synthetic biological circuits incorporating optogenetics, where multiple circuit components can be controlled using designer light induction programs, with broad implications for biotechnology and bioengineering.


Subject(s)
Bayes Theorem , Optogenetics , Saccharomyces cerevisiae , Optogenetics/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Synthetic Biology/methods , Light , Transcription Factors/metabolism , Transcription Factors/genetics , Machine Learning , High-Throughput Screening Assays/methods
15.
Nat Commun ; 15(1): 3525, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664445

ABSTRACT

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.


Subject(s)
Hydrogels , Mice, Transgenic , Optogenetics , Polymers , Polyvinyl Alcohol , Animals , Polyvinyl Alcohol/chemistry , Mice , Hydrogels/chemistry , Optogenetics/methods , Polymers/chemistry , Nanotubes, Carbon/chemistry , Ventral Tegmental Area/physiology , Microelectrodes , Male , Channelrhodopsins/metabolism , Channelrhodopsins/chemistry , Channelrhodopsins/genetics
16.
PLoS Biol ; 22(4): e3002591, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652732

ABSTRACT

Lysosomes are degradation centers of cells and intracellular hubs of signal transduction, nutrient sensing, and autophagy regulation. Dysfunction of lysosomes contributes to a variety of diseases, such as lysosomal storage diseases (LSDs) and neurodegeneration, but the mechanisms are not well understood. Altering lysosomal activity and examining its impact on the occurrence and development of disease is an important strategy for studying lysosome-related diseases. However, methods to dynamically regulate lysosomal function in living cells or animals are still lacking. Here, we constructed lysosome-localized optogenetic actuators, named lyso-NpHR3.0, lyso-ArchT, and lyso-ChR2, to achieve optogenetic manipulation of lysosomes. These new actuators enable light-dependent control of lysosomal membrane potential, pH, hydrolase activity, degradation, and Ca2+ dynamics in living cells. Notably, lyso-ChR2 activation induces autophagy through the mTOR pathway, promotes Aß clearance in an autophagy-dependent manner in cellular models, and alleviates Aß-induced paralysis in the Caenorhabditis elegans model of Alzheimer's disease. Our lysosomal optogenetic actuators supplement the optogenetic toolbox and provide a method to dynamically regulate lysosomal physiology and function in living cells and animals.


Subject(s)
Amyloid beta-Peptides , Autophagy , Caenorhabditis elegans , Lysosomes , Optogenetics , Lysosomes/metabolism , Autophagy/physiology , Optogenetics/methods , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Amyloid beta-Peptides/metabolism , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Calcium/metabolism , TOR Serine-Threonine Kinases/metabolism , Hydrogen-Ion Concentration , HEK293 Cells , HeLa Cells
17.
World J Gastroenterol ; 30(13): 1780-1790, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38659489

ABSTRACT

Colorectal cancer (CRC) has remained the second and the third leading cause of cancer-related death worldwide and in the United States, respectively. Although significant improvement in overall survival has been achieved, death in adult populations under the age of 55 appears to have increased in the past decades. Although new classes of therapeutic strategies such as immunotherapy have emerged, their application is very limited in CRC so far. Microtubule (MT) inhibitors such as taxanes, are not generally successful in CRC. There may be some way to make MT inhibitors work effectively in CRC. One potential advantage that we can take to treat CRC may be the combination of optical techniques coupled to an endoscope or other fiber optics-based devices. A combination of optical devices and photo-activatable drugs may allow us to locally target advanced CRC cells with highly potent MT-targeting drugs. In this Editorial review, we would like to discuss the potential of optogenetic approaches in CRC management.


Subject(s)
Colorectal Neoplasms , Microtubules , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/therapy , Humans , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Optogenetics/methods , Tubulin Modulators/therapeutic use , Tubulin Modulators/pharmacology
18.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38658139

ABSTRACT

High-density linear probes, such as Neuropixels, provide an unprecedented opportunity to understand how neural populations within specific laminar compartments contribute to behavior. Marmoset monkeys, unlike macaque monkeys, have a lissencephalic (smooth) cortex that enables recording perpendicular to the cortical surface, thus making them an ideal animal model for studying laminar computations. Here we present a method for acute Neuropixels recordings in the common marmoset (Callithrix jacchus). The approach replaces the native dura with an artificial silicon-based dura that grants visual access to the cortical surface, which is helpful in avoiding blood vessels, ensures perpendicular penetrations, and could be used in conjunction with optical imaging or optogenetic techniques. The chamber housing the artificial dura is simple to maintain with minimal risk of infection and could be combined with semichronic microdrives and wireless recording hardware. This technique enables repeated acute penetrations over a period of several months. With occasional removal of tissue growth on the pial surface, recordings can be performed for a year or more. The approach is fully compatible with Neuropixels probes, enabling the recording of hundreds of single neurons distributed throughout the cortical column.


Subject(s)
Callithrix , Animals , Dura Mater/physiology , Neurons/physiology , Male , Female , Electrodes, Implanted , Cerebral Cortex/physiology , Optogenetics/methods
19.
Curr Gene Ther ; 24(3): 208-216, 2024.
Article in English | MEDLINE | ID: mdl-38676313

ABSTRACT

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.


Subject(s)
Auditory Perception , Genetic Therapy , Genetic Vectors , Hearing Loss , Optogenetics , Optogenetics/methods , Humans , Genetic Therapy/methods , Auditory Perception/genetics , Genetic Vectors/genetics , Hearing Loss/genetics , Hearing Loss/therapy , Channelrhodopsins/genetics , Dependovirus/genetics , Gene Transfer Techniques , Animals , Cochlear Implants
20.
Nat Commun ; 15(1): 3480, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658537

ABSTRACT

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.


Subject(s)
Caenorhabditis elegans , Neurons , Optogenetics , Zebrafish , Animals , Caenorhabditis elegans/genetics , Neurons/metabolism , Neurons/physiology , Optogenetics/methods , Channelrhodopsins/metabolism , Channelrhodopsins/genetics , Humans , Drosophila , Potassium Channels/metabolism , Potassium Channels/genetics , Chlorides/metabolism , Animals, Genetically Modified , Behavior, Animal , HEK293 Cells , Drosophila melanogaster
SELECTION OF CITATIONS
SEARCH DETAIL
...