Main Menu (Mobile)- Block

Main Menu - Block

janelia7_blocks-janelia7_fake_breadcrumb | block
Huston Lab / Publications
custom | custom

Filter

facetapi-Q2b17qCsTdECvJIqZJgYMaGsr8vANl1n | block

Associated Lab

facetapi-PV5lg7xuz68EAY8eakJzrcmwtdGEnxR0 | block
facetapi-021SKYQnqXW6ODq5W5dPAFEDBaEJubhN | block
general_search_page-panel_pane_1 | views_panes

9 Publications

Showing 1-9 of 9 results
Your Criteria:
    03/13/24 | Carbon Nanomaterial Fluorescent Probes and Their Biological Applications
    Krasley AT, Li E, Galeana JM, Bulumulla C, Beyene AG, Demirer GS
    Chemical Reviews. 2024-03-13:. doi: 10.1021/acs.chemrev.3c00581

    Fluorescent carbon nanomaterials have broadly useful chemical and photophysical attributes that are conducive to applications in biology. In this review, we focus on materials whose photophysics allow for the use of these materials in biomedical and environmental applications, with emphasis on imaging, biosensing, and cargo delivery. The review focuses primarily on graphitic carbon nanomaterials including graphene and its derivatives, carbon nanotubes, as well as carbon dots and carbon nanohoops. Recent advances in and future prospects of these fields are discussed at depth, and where appropriate, references to reviews pertaining to older literature are provided.

    View Publication Page
    07/04/22 | Visualizing Synaptic Dopamine Efflux with a 2D Nanofilm.
    Chandima Bulumulla , Andrew T. Krasley , Deepika Walpita , Abraham G. Beyene
    eLife. 2022 Jul 04:. doi: 10.7554/eLife.78773

    Chemical neurotransmission constitutes one of the fundamental modalities of communication between neurons. Monitoring release of these chemicals has traditionally been difficult to carry out at spatial and temporal scales relevant to neuron function. To understand chemical neurotransmission more fully, we need to improve the spatial and temporal resolutions of measurements for neurotransmitter release. To address this, we engineered a chemi-sensitive, two-dimensional nanofilm that facilitates subcellular visualization of the release and diffusion of the neurochemical dopamine with synaptic resolution, quantal sensitivity, and simultaneously from hundreds of release sites. Using this technology, we were able to monitor the spatiotemporal dynamics of dopamine release in dendritic processes, a poorly understood phenomenon. We found that dopamine release is broadcast from a subset of dendritic processes as hotspots that have a mean spatial spread of ≈3.2 µm (full width at half maximum) and are observed with a mean spatial frequency of 1 hotspot per ≈7.5 µm of dendritic length. Major dendrites of dopamine neurons and fine dendritic processes, as well as dendritic arbors and dendrites with no apparent varicose morphology participated in dopamine release. Remarkably, these release hotspots colocalized with Bassoon, suggesting that Bassoon may contribute to organizing active zones in dendrites, similar to its role in axon terminals.

    View Publication Page
    05/21/21 | Near-infrared catecholamine nanosensors for high spatiotemporal dopamine imaging.
    Yang SJ, Del Bonis-O'Donnell JT, Beyene AG, Landry MP
    Nature Protocols. 2021 May 21;16(6):3026-3048. doi: 10.1038/s41596-021-00530-4

    Dopamine neuromodulation of neural synapses is a process implicated in a number of critical brain functions and diseases. Development of protocols to visualize this dynamic neurochemical process is essential to understanding how dopamine modulates brain function. We have developed a non-genetically encoded, near-IR (nIR) catecholamine nanosensor (nIRCat) capable of identifying ~2-µm dopamine release hotspots in dorsal striatal brain slices. nIRCat is readily synthesized through sonication of single walled carbon nanotubes with DNA oligos, can be readily introduced into both genetically tractable and intractable organisms and is compatible with a number of dopamine receptor agonists and antagonists. Here we describe the synthesis, characterization and implementation of nIRCat in acute mouse brain slices. We demonstrate how nIRCat can be used to image electrically or optogenetically stimulated dopamine release, and how these procedures can be leveraged to study the effects of dopamine receptor pharmacology. In addition, we provide suggestions for building or adapting wide-field microscopy to be compatible with nIRCat nIR fluorescence imaging. We discuss strategies for analyzing nIR video data to identify dopamine release hotspots and quantify their kinetics. This protocol can be adapted and implemented for imaging other neuromodulators by using probes of this class and can be used in a broad range of species without genetic manipulation. The synthesis and characterization protocols for nIRCat take ~5 h, and the preparation and fluorescence imaging of live brain slices by using nIRCats require ~6 h.

    View Publication Page
    11/01/20 | A community for Black chemists.
    Beyene AG, Panescu P
    Nature Chemistry. 2020 Nov 01;12(11):988-989. doi: 10.1038/s41557-020-00572-3
    07/10/19 | Imaging striatal dopamine release using a nongenetically encoded near infrared fluorescent catecholamine nanosensor.
    Beyene AG, Delevich K, Del Bonis-O’Donnell JT, Piekarski DJ, Lin WC, Thomas AW, Yang SJ, Kosillo P, Yang D, Prounis GS, Wilbrecht L, Landry MP
    Science Advances. 2019 Jul 10;5(7):eaaw3108. doi: 10.1126/sciadv.aaw3108

    Neuromodulation plays a critical role in brain function in both health and disease, and new tools that capture neuromodulation with high spatial and temporal resolution are needed. Here, we introduce a synthetic catecholamine nanosensor with fluorescent emission in the near infrared range (1000–1300 nm), near infrared catecholamine nanosensor (nIRCat). We demonstrate that nIRCats can be used to measure electrically and optogenetically evoked dopamine release in brain tissue, revealing hotspots with a median size of 2 µm. We also demonstrated that nIRCats are compatible with dopamine pharmacology and show D2 autoreceptor modulation of evoked dopamine release, which varied as a function of initial release magnitude at different hotspots. Together, our data demonstrate that nIRCats and other nanosensors of this class can serve as versatile synthetic optical tools to monitor neuromodulatory neurotransmitter release with high spatial resolution.

    View Publication Page
    11/14/18 | Ultralarge modulation of fluorescence by neuromodulators in carbon nanotubes functionalized with self-assembled oligonucleotide rings.
    Beyene AG, Alizadehmojarad AA, Dorlhiac G, Goh N, Streets AM, Král P, Vuković L, Landry MP
    Nano Letters. 2018 Nov 14;18(11):6995-7003. doi: 10.1021/acs.nanolett.8b02937

    Noncovalent interactions between single-stranded DNA (ssDNA) oligonucleotides and single wall carbon nanotubes (SWNTs) have provided a unique class of tunable chemistries for a variety of applications. However, mechanistic insight into both the photophysical and intermolecular phenomena underlying their utility is lacking, which results in obligate heuristic approaches for producing ssDNA-SWNT based technologies. In this work, we present an ultrasensitive "turn-on" nanosensor for neuromodulators dopamine and norepinephrine with strong relative change in fluorescence intensity (Δ F/ F) of up to 3500%, a signal appropriate for in vivo neuroimaging, and uncover the photophysical principles and intermolecular interactions that govern the molecular recognition and fluorescence modulation of this nanosensor synthesized from the spontaneous self-assembly of (GT) ssDNA rings on SWNTs. The fluorescence modulation of the ssDNA-SWNT conjugate is shown to exhibit remarkable sensitivity to the ssDNA sequence chemistry, length, and surface density, providing a set of parameters with which to tune nanosensor dynamic range, analyte selectivity and strength of fluorescence turn-on. We employ classical and quantum mechanical molecular dynamics simulations to rationalize our experimental findings. Calculations show that (GT) ssDNA form ordered rings around (9,4) SWNTs, inducing periodic surface potentials that modulate exciton recombination lifetimes. Further evidence is presented to elucidate how dopamine analyte binding modulates SWNT fluorescence. We discuss the implications of our findings for SWNT-based molecular imaging applications.

    View Publication Page
    11/13/18 | New optical probes bring dopamine to light.
    Beyene AG, Delevich K, Yang SJ, Landry MP
    Biochemistry. 2018 Nov 13;57(45):6379-6381. doi: 10.1021/acs.biochem.8b00883

    Chemical signaling between neurons in the brain can be divided into two major categories: fast synaptic transmission and neuromodulation. Fast synaptic transmission, mediated by amino acids such as glutamate and GABA, occurs on millisecond time scales and results in the influx of ions through ligand-gated ion channels on postsynaptic neurons (Figure 1A). Electrophysiological and optical imaging tools, including genetically encoded voltage indicators, have enabled neuroscientists to link cause (neurotransmitter release) and effect (membrane polarization) of synaptic transmission in time and space. Unlike classical neurotransmitters, neuromodulators do not produce immediate electrical effects that excite or inhibit target neurons. Instead, neuromodulators tune the intrinsic or synaptic properties of neurons, most commonly through interaction with G-protein-coupled receptors (GPCRs) (Figure 1B). Neuromodulators can escape the synaptic cleft and diffuse broadly, allowing them to influence the activity of many neurons in a state-dependent manner. Therefore, the spatial component of neuromodulator flux is fundamentally important. However, the temporal and/or spatial limitations of techniques classically used to study neuromodulation, such as microdialysis and fast-scan cyclic voltammetry (FSCV), make it difficult to interpret how neuromodulator release affects the plasticity or function of target neuronal populations on a moment-to-moment basis. Therefore, tools that can detect neuromodulators with high spatiotemporal resolution are critical for understanding their impact on neural computations that control behavior in health and disease.

    View Publication Page
    10/18/17 | Stochastic simulation of dopamine neuromodulation for implementation of fluorescent neurochemical probes in the striatal extracellular space.
    Beyene AG, McFarlane IR, Pinals RL, Landry MP
    ACS Chemical Neuroscience. 2017 Oct 18;8(10):2275-2289. doi: 10.1021/acschemneuro.7b00193

    Imaging the dynamic behavior of neuromodulatory neurotransmitters in the extracelluar space that arise from individual quantal release events would constitute a major advance in neurochemical imaging. Spatial and temporal resolution of these highly stochastic neuromodulatory events requires concurrent advances in the chemical development of optical nanosensors selective for neuromodulators in concert with advances in imaging methodologies to capture millisecond neurotransmitter release. Herein, we develop and implement a stochastic model to describe dopamine dynamics in the extracellular space (ECS) of the brain dorsal striatum to guide the design and implementation of fluorescent neurochemical probes that record neurotransmitter dynamics in the ECS. Our model is developed from first-principles and simulates release, diffusion, and reuptake of dopamine in a 3D simulation volume of striatal tissue. We find that in vivo imaging of neuromodulation requires simultaneous optimization of dopamine nanosensor reversibility and sensitivity: dopamine imaging in the striatum or nucleus accumbens requires nanosensors with an optimal dopamine dissociation constant (K) of 1 μM, whereas Ks above 10 μM are required for dopamine imaging in the prefrontal cortex. Furthermore, as a result of the probabilistic nature of dopamine terminal activity in the striatum, our model reveals that imaging frame rates of 20 Hz are optimal for recording temporally resolved dopamine release events. Our work provides a modeling platform to probe how complex neuromodulatory processes can be studied with fluorescent nanosensors and enables direct evaluation of nanosensor chemistry and imaging hardware parameters. Our stochastic model is generic for evaluating fluorescent neurotransmission probes, and is broadly applicable to the design of other neurotransmitter fluorophores and their optimization for implementation in vivo.

    View Publication Page
    08/15/17 | Dual near-Infrared two-photon microscopy for deep-tissue dopamine nanosensor imaging.
    Bonis-O'Donnell JT, Page RH, Beyene AG, Tindall EG, McFarlane IR, Landry MP
    Advanced Functional Materials. 2017 August 15;27(39):1702112. doi: 10.1002/adfm.v27.3910.1002/adfm.201702112

    A key limitation for achieving deep imaging in biological structures lies in photon absorption and scattering leading to attenuation of fluorescence. In particular, neurotransmitter imaging is challenging in the biologically relevant context of the intact brain for which photons must traverse the cranium, skin, and bone. Thus, fluorescence imaging is limited to the surface cortical layers of the brain, only achievable with craniotomy. Herein, this study describes optimal excitation and emission wavelengths for through‐cranium imaging, and demonstrates that near‐infrared emissive nanosensors can be photoexcited using a two‐photon 1560 nm excitation source. Dopamine‐sensitive nanosensors can undergo two‐photon excitation, and provide chirality‐dependent responses selective for dopamine with fluorescent turn‐on responses varying between 20% and 350%. The two‐photon absorption cross‐section and quantum yield of dopamine nanosensors are further calculated, and a two‐photon power law relationship for the nanosensor excitation process is confirmed. Finally, the improved image quality of the nanosensors embedded 2‐mm‐deep into a brain‐mimetic tissue phantom is shown, whereby one‐photon excitation yields 42% scattering, in contrast to 4% scattering when the same object is imaged under two‐photon excitation. The approach overcomes traditional limitations in deep‐tissue fluorescence microscopy, and can enable neurotransmitter imaging in the biologically relevant milieu of the intact and living brain.

    View Publication Page