1
|
Zabelskii D, Bukhdruker S, Bukhalovich S, Tsybrov F, Lamm GHU, Astashkin R, Doroginin D, Matveev G, Sudarev V, Kuzmin A, Zinovev E, Vlasova A, Ryzhykau Y, Ilyinsky N, Gushchin I, Bourenkov G, Alekseev A, Round A, Wachtveitl J, Bamberg E, Gordeliy V. Ion-conducting and gating molecular mechanisms of channelrhodopsin revealed by true-atomic-resolution structures of open and closed states. Nat Struct Mol Biol 2025:10.1038/s41594-025-01488-7. [PMID: 40205223 DOI: 10.1038/s41594-025-01488-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/09/2025] [Indexed: 04/11/2025]
Abstract
Channelrhodopsins (ChRs) have emerged as major optogenetics tools, particularly in neuroscience. Despite their importance, the molecular mechanism of ChR opening remains elusive. Moreover, all reported structures of ChRs correspond to either a closed or an early intermediate state and lack the necessary level of detail owing to the limited resolution. Here we present the structures of the closed and open states of a cation-conducting ChR, OLPVR1, from Organic Lake phycodnavirus, belonging to the family of viral ChRs solved at 1.1- and 1.3-Å resolution at physiologically relevant pH conditions (pH 8.0). OLPVR1 was expressed in Escherichia coli and crystallized using an in meso approach, and the structures were solved by X-ray crystallography. We also present the structure of the OLPVR1 protonated state at acidic pH (pH 2.5) at 1.4-Å resolution. Together, these three structures elucidate the molecular mechanisms of the channel's opening and permeability in detail. Extensive functional studies support the proposed mechanisms. Channel opening is controlled by isomerization of the retinal cofactor, triggering protonation of proton acceptors and deprotonation of proton donors located in the three gates of the channel. The E51 residue in the core of the central gate (similar to E90 of ChR2 from Chlamydomonas reinhardtii) plays a key role in the opening of the channel. E51 flips out of the gate and towards the proton acceptor D200 (D253 in ChR2 in C. reinhardtii), establishing a hydrogen bond between them. Despite differences in subfamilies of ChRs, they share a common gate-cavity architecture, suggesting that they could have similar general gating mechanisms. These results enabled us to design viral rhodopsin with improved properties for optogenetic applications. The structural data and mechanisms might also be helpful for better understanding other ChRs and their engineering.
Collapse
Affiliation(s)
| | - Sergey Bukhdruker
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Siarhei Bukhalovich
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Fedor Tsybrov
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Gerrit H U Lamm
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Roman Astashkin
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Demid Doroginin
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Grigory Matveev
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Vsevolod Sudarev
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Alexander Kuzmin
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Egor Zinovev
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Anastasiia Vlasova
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Yury Ryzhykau
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Dubna, Russia
| | - Nikolay Ilyinsky
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Ivan Gushchin
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Gleb Bourenkov
- European Molecular Biology Laboratory, Hamburg unit c/o DESY, Hamburg, Germany
| | - Alexey Alekseev
- Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Adam Round
- European X-ray Free Electron Laser GmbH, Schenefeld, Germany
| | - Josef Wachtveitl
- Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ernst Bamberg
- Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| | - Valentin Gordeliy
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France.
| |
Collapse
|
2
|
Zhang X, Dhir S, Melidis L, Chen Y, Yu Z, Simeone A, Spiegel J, Adhikari S, Balasubramanian S. Optical control of gene expression using a DNA G-quadruplex targeting reversible photoswitch. Nat Chem 2025:10.1038/s41557-025-01792-1. [PMID: 40181150 DOI: 10.1038/s41557-025-01792-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025]
Abstract
Transcriptional regulation is a dynamic process that coordinates diverse cellular activities, and the use of small molecules to perturb gene expression has propelled our understanding of the fundamental regulatory mechanisms. However, small molecules typically lack the spatiotemporal precision required in highly non-invasive, controlled settings. Here we present the development of a cell-permeable small-molecule DNA G-quadruplex (G4) binder, termed G4switch, that can be reversibly toggled on and off by visible light. We have biophysically characterized the light-mediated control of G4 binding in vitro, followed by cellular, genomic mapping of G4switch to G4 targets in chromatin to confirm G4-selective, light-dependent binding in a cellular context. By deploying G4switch in living cells, we show spatiotemporal control over the expression of a set of G4-containing genes and G4-associated cell proliferation. Our studies demonstrate a chemical tool and approach to interrogate the dynamics of key biological processes directly at the molecular level in cells.
Collapse
Affiliation(s)
- Xiaoyun Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Somdutta Dhir
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Larry Melidis
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Yuqi Chen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Zutao Yu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Angela Simeone
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Jochen Spiegel
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Santosh Adhikari
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Shankar Balasubramanian
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK.
- School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Schad C, Ray C, Díaz-Norambuena C, Serrano-Buitrago S, Moreno F, Maroto BL, García-Moreno I, Muñoz-Úbeda M, López-Montero I, Bañuelos J, de la Moya S. Water-soluble BODIPY dyes: a novel approach for their sustainable chemistry and applied photonics. Chem Sci 2025:d5sc01295c. [PMID: 40206553 PMCID: PMC11976447 DOI: 10.1039/d5sc01295c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
The BODIPY family of organic dyes has emerged as a cornerstone in photonics research development, driving innovation and advancement in various fields of high socio-economic interest. However, the majority of BODIPY dyes exhibit hydrophobic characteristics, resulting in poor solubility in water and other hydrophilic solvents. This solubility is paramount for their optimal utilization in a myriad of photonic applications, particularly in the realms of biology and medicine. Furthermore, it facilitates safer and more sustainable manipulation and chemical modification of these expansive dyes. Nevertheless, bestowing BODIPYs with water solubility while preserving their other essential properties, notably their photophysical signatures, poses a significant challenge. In this context, we present a straightforward general chemical modification aimed at converting conventional hydrophobic BODIPYs into highly hydrophilic variants, thus enabling their efficient solubilization in water and other hydrophilic solvents with minimal disruption to the dye's inherent photophysics. The efficacy of this methodology is demonstrated through the synthesis of a number of water-soluble BODIPY dyes featuring diverse substitution patterns. Furthermore, we showcase their utility in a spectrum of photonics-related applications, including in-water BODIPY chemistry and dye-laser technology, and fluorescence microscopy.
Collapse
Affiliation(s)
- Christopher Schad
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
| | - Cesar Ray
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
| | - Carolina Díaz-Norambuena
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
- Departamento de Química-Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco-EHU Bilbao 48080 Spain
| | - Sergio Serrano-Buitrago
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
| | - Florencio Moreno
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
| | - Beatriz L Maroto
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
| | - Inmaculada García-Moreno
- Departamento de Química-Física de Materiales, Instituto de Química-Física Blas Cabrera, Consejo Superior de Investigaciones Científicas (CSIC) Serrano 119 Madrid 28006 Spain
| | - Mónica Muñoz-Úbeda
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n 28040 Madrid Spain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12) Avda. de Córdoba s/n 28041 Madrid Spain
| | - Iván López-Montero
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n 28040 Madrid Spain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12) Avda. de Córdoba s/n 28041 Madrid Spain
- Instituto Pluridisciplinar, Universidad Complutense de Madrid P° Juan XXIII 1 28040 Madrid Spain
| | - Jorge Bañuelos
- Departamento de Química-Física, Facultad de Ciencia y Tecnología, Universidad del País Vasco-EHU Bilbao 48080 Spain
| | - Santiago de la Moya
- Departamento de Química Orgánica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Ciudad Universitaria s/n Madrid 28040 Spain
| |
Collapse
|
4
|
Hsu LM, Shih YYI. Neuromodulation in Small Animal fMRI. J Magn Reson Imaging 2025; 61:1597-1617. [PMID: 39279265 PMCID: PMC11903207 DOI: 10.1002/jmri.29575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/18/2024] Open
Abstract
The integration of functional magnetic resonance imaging (fMRI) with advanced neuroscience technologies in experimental small animal models offers a unique path to interrogate the causal relationships between regional brain activity and brain-wide network measures-a goal challenging to accomplish in human subjects. This review traces the historical development of the neuromodulation techniques commonly used in rodents, such as electrical deep brain stimulation, optogenetics, and chemogenetics, and focuses on their application with fMRI. We discuss their advantageousness roles in uncovering the signaling architecture within the brain and the methodological considerations necessary when conducting these experiments. By presenting several rodent-based case studies, we aim to demonstrate the potential of the multimodal neuromodulation approach in shedding light on neurovascular coupling, the neural basis of brain network functions, and their connections to behaviors. Key findings highlight the cell-type and circuit-specific modulation of brain-wide activity patterns and their behavioral correlates. We also discuss several future directions and feature the use of mediation and moderation analytical models beyond the intuitive evoked response mapping, to better leverage the rich information available in fMRI data with neuromodulation. Using fMRI alongside neuromodulation techniques provide insights into the mesoscopic (relating to the intermediate scale between single neurons and large-scale brain networks) and macroscopic fMRI measures that correlate with specific neuronal events. This integration bridges the gap between different scales of neuroscience research, facilitating the exploration and testing of novel therapeutic strategies aimed at altering network-mediated behaviors. In conclusion, the combination of fMRI with neuromodulation techniques provides crucial insights into mesoscopic and macroscopic brain dynamics, advancing our understanding of brain function in health and disease. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Li-Ming Hsu
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Radiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yen-Yu Ian Shih
- Center for Animal Magnetic Resonance Imaging, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Neurology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Jeong D, Kim S, Park H, Woo K, Choi J, Choi M, Shin J, Park SH, Seon M, Lee D, Cha J, Kim Y. Optogenetically Activatable MLKL as a Standalone Functional Module for Necroptosis and Therapeutic Applications in Antitumoral Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412393. [PMID: 39921454 PMCID: PMC11967802 DOI: 10.1002/advs.202412393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/17/2025] [Indexed: 02/10/2025]
Abstract
Necroptosis plays a crucial role in the progression of various diseases and has gained substantial attention for its potential to activate antitumor immunity. However, the complex signaling networks that regulate necroptosis have made it challenging to fully understand its mechanisms and translate this knowledge into therapeutic applications. To address these challenges, an optogenetically activatable necroptosis system is developed that allows for precise spatiotemporal control of key necroptosis regulators, bypassing complex upstream signaling processes. The system, specifically featuring optoMLKL, demonstrates that it can rapidly assemble into functional higher-order "hotspots" within cellular membrane compartments, independent of RIPK3-mediated phosphorylation. Moreover, the functional module of optoMLKL significantly enhances innate immune responses by promoting the release of iDAMPs and cDAMPs, which are critical for initiating antitumor immunity. Furthermore, optoMLKL exhibits antitumor effects when activated in patient-derived pancreatic cancer organoids, highlighting its potential for clinical application. These findings will pave the way for innovative cancer therapies by leveraging optogenetic approaches to precisely control and enhance necroptosis.
Collapse
Affiliation(s)
- Da‐Hye Jeong
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Seokhwi Kim
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Han‐Hee Park
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Kyoung‐Jin Woo
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Jae‐Il Choi
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Minji Choi
- Program in Biomedical Science and EngineeringGraduate schoolInha UniversityIncheon22212Republic of Korea
| | - Jisoo Shin
- Program in Biomedical Science and EngineeringGraduate schoolInha UniversityIncheon22212Republic of Korea
| | - So Hyun Park
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Myung‐Wook Seon
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| | - Dakeun Lee
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
- Department of PathologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Jong‐Ho Cha
- Program in Biomedical Science and EngineeringGraduate schoolInha UniversityIncheon22212Republic of Korea
- Department of Biomedical SciencesCollege of MedicineInha UniversityIncheon22212Republic of Korea
- Biohybrid Systems Research CenterInha UniversityIncheon22212Republic of Korea
| | - You‐Sun Kim
- Department of BiochemistryAjou University School of MedicineSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate School of Ajou UniversitySuwon16499Republic of Korea
| |
Collapse
|
6
|
Dixit N, Pyari G, Bansal H, Roy S. Theoretical analysis of low power optogenetic control of synaptic plasticity with subcellular expression of CapChR2 at postsynaptic spine. Sci Rep 2025; 15:11166. [PMID: 40169824 PMCID: PMC11962105 DOI: 10.1038/s41598-025-95355-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
Precise control of intracellular calcium ([Formula: see text]) concentration at the synaptic neuron terminal can unravel the mechanism behind computation, learning, and memory formation inside the brain. Recently, the discovery of [Formula: see text]-permeable channelrhodopsins (CapChRs) has opened the opportunity to effectively control the intracellular [Formula: see text] concentration using optogenetics. Here, we present a new theoretical model for precise optogenetic control with newly discovered CapChR2 at postsynaptic neuron. A detailed theoretical analysis of coincident stimulation of presynaptic terminal, postsynaptic spine and optogenetic activation of CapChR2-expressing postsynaptic spine shows different ways to control postsynaptic intracellular [Formula: see text] concentration. Irradiance-dependent [Formula: see text] flow is an additional advantage of this novel method. The minimum threshold of light irradiance and optimal ranges of time lag among different stimulations and stimulation frequencies have also been determined. It is shown that synaptic efficacy occurs at 20 µW/mm2 at coincident electrical stimulation of presynaptic terminal and postsynaptic spine with optogenetic activation of CapChR2-expressed postsynaptic spine. The analysis provides a new means of direct optogenetic control of [Formula: see text]-based synaptic plasticity, better understanding of learning and memory processes, and opens prospects for targeted therapeutic interventions to modulate synaptic function and address various neurological disorders.
Collapse
Affiliation(s)
- Nripesh Dixit
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India
| | - Gur Pyari
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India
| | - Himanshu Bansal
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India
| | - Sukhdev Roy
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, 282005, India.
| |
Collapse
|
7
|
Renteria CA, Kahng J, Tibble B, Iyer RR, Shi J, Algrain H, Chaney EJ, Aksamitiene E, Liu YZ, Robinson P, Schmidt T, Boppart SA. Two-photon activation, deactivation, and coherent control of melanopsin in live cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645437. [PMID: 40196647 PMCID: PMC11974792 DOI: 10.1101/2025.03.26.645437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intrinsically photosensitive retinal ganglion cells are photoreceptors discovered in the last 20 years. These cells project to the suprachiasmatic nucleus of the brain to drive circadian rhythms, regulated by ambient light levels. The photopigment responsible for photoactivation in these cells, melanopsin, has been shown to exhibit many unique activation features among opsins. Notably, the photopigment can exist in three states dependent on the intensity and spectrum of ambient light, which affects its function. Despite increasing knowledge about these cells and melanopsin, tools that can manipulate their three states, and do so with single-cell precision, are limited. This reduces the extent to which circuit-level phenomena, and studying the implications of melanopsin tri-stability in living systems, can be pursued. In this report, we evoke and modulate calcium transients in live cells and intrinsically photosensitive retinal ganglion cells from isolated retinal tissues following two-photon excitation using near-infrared light pulses. We demonstrate that two-photon activation of melanopsin can successfully stimulate melanopsin-expressing cells with high spatio-temporal precision. Moreover, we demonstrate that the functional tri-stability of the photopigment can be interrogated by multiphoton excitation using spectral-temporal modulation of a broadband, ultrafast laser source.
Collapse
|
8
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
9
|
Ovechkina VS, Andrianova SK, Shimanskaia IO, Suvorova PS, Ryabinina AY, Blagonravov ML, Belousov VV, Mozhaev AA. Advances in Optogenetics and Thermogenetics for Control of Non-Neuronal Cells and Tissues in Biomedical Research. ACS Chem Biol 2025; 20:553-572. [PMID: 40056098 DOI: 10.1021/acschembio.4c00842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Optogenetics and chemogenetics are relatively new biomedical technologies that emerged 20 years ago and have been evolving rapidly since then. This has been made possible by the combined use of genetic engineering, optics, and electrophysiology. With the development of optogenetics and thermogenetics, the molecular tools for cellular control are continuously being optimized, studied, and modified, expanding both their applications and their biomedical uses. The most notable changes have occurred in the basic life sciences, especially in neurobiology and the activation of neurons to control behavior. Currently, these methods of activation have gone far beyond neurobiology and are being used in cardiovascular research, for potential cancer therapy, to control metabolism, etc. In this review, we provide brief information on the types of molecular tools for optogenetic and thermogenetic methods─microbial rhodopsins and proteins of the TRP superfamily─and also consider their applications in the field of activation of non-neuronal tissues and mammalian cells. We also consider the potential of these technologies and the prospects for the use of optogenetics and thermogenetics in biomedical research.
Collapse
Affiliation(s)
- Vera S Ovechkina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Sofya K Andrianova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Iana O Shimanskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Polina S Suvorova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Anna Y Ryabinina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Mikhail L Blagonravov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Vsevolod V Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117513, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, 121205, Russia
| | - Andrey A Mozhaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| |
Collapse
|
10
|
Huang J, Xue S, Teixeira AP, Fussenegger M. A mediator-free sonogenetic switch for therapeutic protein expression in mammalian cells. Nucleic Acids Res 2025; 53:gkaf191. [PMID: 40114374 PMCID: PMC11925730 DOI: 10.1093/nar/gkaf191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/28/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025] Open
Abstract
An ultrasound-responsive transgene circuit can provide non-invasive, spatiotemporally precise remote control of gene expression and cellular behavior in synthetic biology applications. However, current ultrasound-based systems often rely on nanoparticles or harness ultrasound's thermal effects, posing risks of tissue damage and cellular stress that limit their therapeutic potential. Here, we present Spatiotemporal Ultrasound-induced Protein Expression Regulator (SUPER), a novel gene switch enabling mediator-free, non-invasive and direct regulation of protein expression via ultrasound in mammalian cells. SUPER leverages the mammalian reactive oxygen species (ROS) sensing system, featuring KEAP1 (Kelch-like ECH-associated protein 1), NRF2 (nuclear factor erythroid 2-related factor 2), and antioxidant response element (ARE) as its core components. We demonstrate that low-intensity (1.5 W/cm2, ∼45 kHz), brief (40 s) ultrasound exposure generates non-toxic levels of ROS, activating the KEAP1/NRF2 pathway in engineered cells and leading to the controlled expression of target gene(s) via a synthetic ARE promoter. The system exhibits robust expression dynamics, excellent reversibility, and functionality in various cell types, including human mesenchymal stem cell-derived lines (hMSC-TERT). In a proof-of-concept study, ultrasound stimulation of subcutaneously implanted microencapsulated engineered cells stably expressing the sonogenetic circuit in a type 1 diabetic mouse model triggered sufficient insulin production to restore normoglycemia. Our work highlights ultrasound's potential as a precise and non-invasive tool for advancing cell and gene therapies in personalized medicine.
Collapse
Affiliation(s)
- Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| | - Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
- Faculty of Science, University of Basel, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| |
Collapse
|
11
|
Duffy JS, Bellgrove MA, Murphy PR, O'Connell RG. Disentangling sources of variability in decision-making. Nat Rev Neurosci 2025:10.1038/s41583-025-00916-3. [PMID: 40114010 DOI: 10.1038/s41583-025-00916-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Even the most highly-trained observers presented with identical choice-relevant stimuli will reliably exhibit substantial trial-to-trial variability in the timing and accuracy of their choices. Despite being a pervasive feature of choice behaviour and a prominent phenotype for numerous clinical disorders, the capability to disentangle the sources of such intra-individual variability (IIV) remains limited. In principle, computational models of decision-making offer a means of parsing and estimating these sources, but methodological limitations have prevented this potential from being fully realized in practice. In this Review, we first discuss current limitations of algorithmic models for understanding variability in decision-making behaviour. We then highlight recent advances in behavioural paradigm design, novel analyses of cross-trial behavioural and neural dynamics, and the development of neurally grounded computational models that are now making it possible to link distinct components of IIV to well-defined neural processes. Taken together, we demonstrate how these methods are opening up new avenues for systematically analysing the neural origins of IIV, paving the way for a more refined, holistic understanding of decision-making in health and disease.
Collapse
Affiliation(s)
- Jade S Duffy
- Trinity College Institute of Neuroscience and School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Mark A Bellgrove
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
| | - Peter R Murphy
- Department of Psychology, Maynooth University, Kildare, Ireland
| | - Redmond G O'Connell
- Trinity College Institute of Neuroscience and School of Psychology, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
12
|
Hsu LM, Cerri DH, Carelli RM, Shih YYI. Optogenetic stimulation of cell bodies versus axonal terminals generate comparable activity and functional connectivity patterns in the brain. Brain Stimul 2025:S1935-861X(25)00059-2. [PMID: 40090667 DOI: 10.1016/j.brs.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/08/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025] Open
Abstract
Optogenetic techniques are often employed to dissect neural pathways with presumed specificity for targeted projections. In this study, we used optogenetic fMRI to investigate the effective landscape of stimulating the cell bodies versus one of its projection terminals. Specifically, we selected a long-range unidirectional projection from the ventral subiculum (vSUB) to the nucleus accumbens shell (NAcSh) and placed two stimulating fibers-one at the vSUB cell bodies and the other at the vSUB terminals in the NAcSh. Contrary to the conventional view that terminal stimulation confines activity to the feedforward stimulated pathway, our findings reveal that terminal stimulation induces brain activity and connectivity patterns remarkably similar to those of vSUB cell body stimulation. This observation suggests that the specificity of optogenetic terminal stimulation may induce antidromic activation, leading to broader network involvement than previously acknowledged.
Collapse
Affiliation(s)
- Li-Ming Hsu
- Center for Animal MRI, University of North Carolina at Chapel Hill; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill; Department of Radiology, University of North Carolina at Chapel Hill.
| | - Domenic H Cerri
- Center for Animal MRI, University of North Carolina at Chapel Hill; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill; Department of Neurology, University of North Carolina at Chapel Hill
| | - Regina M Carelli
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina at Chapel Hill; Biomedical Research Imaging Center, University of North Carolina at Chapel Hill; Department of Neurology, University of North Carolina at Chapel Hill.
| |
Collapse
|
13
|
Aggarwal S. Recent advances in fundamental research on photon avalanches on the nanometre scale. NANOSCALE 2025; 17:6329-6361. [PMID: 39951321 DOI: 10.1039/d4nr03493g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
In recent years, Photon Avalanche (PA) on the nanometre scale has emerged as a groundbreaking phenomenon, enabling the generation of high-energy photons with minimal pumping power due to its highly nonlinear optical dynamics. This review focuses on the advancement in photon-avalanching nanoparticles (ANPs), composed of lanthanide ion-doped inorganic matrices, which exhibit remarkable optical nonlinear response under low-power excitation. The objective of this article is to provide a comprehensive overview of the PA mechanism in nanoscale materials, with a specific focus on single-ANP systems. Key factors influencing the PA characteristics, such as excitation-power threshold, excited-state absorption, cross-relaxation process, dopant ion concentration, and temperature sensitivity are summarized. Furthermore, the review situates recent ANP research within the broader context of early studies on the PA mechanism observed in bulk crystals and optical fibers, highlighting the distinctive features and applications of ANPs. Notable applications discussed include single-particle and biological super-resolution imaging, deep-tissue imaging, luminescence thermometry, ANP-based lasers, optical data storage, and information security. The paper also addresses current challenges and limitations of ANPs in practical applications, proposing potential solutions and future research directions to facilitate their integration into real-world environments. This review aims to serve as a valuable resource for researchers seeking to advance the understanding and application of ANPs in various scientific and technological domains.
Collapse
Affiliation(s)
- Shradha Aggarwal
- IBS (Institute of Basic Science), 44919, Ulsan, Korea.
- Department of Chemistry, UNIST (Ulsan National Institute of Science and Technology), 44919, Ulsan, Korea
| |
Collapse
|
14
|
Pozo MR, Heinson YW, Chua CJ, Entcheva E. Control of cardiac waves in human iPSC-CM syncytia by a Halbach array and magnetic nanoparticles. Biophys J 2025:S0006-3495(25)00142-0. [PMID: 40077966 DOI: 10.1016/j.bpj.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/11/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
The Halbach array, originally developed for particle accelerators, is a compact arrangement of permanent magnets that creates well-defined magnetic fields without heating. Here, we demonstrate its use for modulating the speed of electromechanical waves in cardiac syncytia of human stem cell-derived cardiomyocytes. At 40-50 mT magnetic field strength, a cylindrical dipolar Halbach array boosted the conduction velocity (CV) by up to 25% when the magnetic field was co-aligned with the electromechanical wave (but not when perpendicular to it). To observe the effects, a short-term incubation of the cardiac cell constructs with non-targeted magnetic nanoparticles (mNPs) was sufficient. This led to increased CV anisotropy, and effects were most pronounced at slower pacing rates. Instantaneous formation and rearrangement of elongated mNP clusters upon magnetic-field rotation was seen, creating dynamic structural anisotropy that may have contributed to the directional CV effects. This approach may be useful for anti-arrhythmic control of cardiac waves.
Collapse
Affiliation(s)
- Maria R Pozo
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Yuli W Heinson
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Christianne J Chua
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, District of Columbia.
| |
Collapse
|
15
|
Hashim PK, Shaji AT, Amrutha AS, Ahmad S. Conceptual expansion of photomedicine for spatiotemporal treatment methods. RSC Med Chem 2025:d4md01005a. [PMID: 40177642 PMCID: PMC11959407 DOI: 10.1039/d4md01005a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Photomedicine has evolved from basic phototherapy to a broad range of light-based technologies to achieve precise and minimally invasive therapeutic outcomes. Recent advances in light sources, photochemical reactions, and photoswitches have facilitated the development of light-activated methodologies for modulating biological processes. This review discusses the history of light therapy that leads to the emergence of a new field known as photopharmacology, mode of actions in photopharmacology such as photodynamic, photo-uncaging and photoswitchable methods, a few representative examples in photopharmacology, and a brief overview of its associated challenges. The current developments in photopharmacology hold great promise for the treatment of diseases such as cancer, with enhanced therapeutic precision, and minimal side effects. We foresee further expansion of photomedicine for novel approaches in precision medicine and healthcare, and unprecedented treatment methods.
Collapse
Affiliation(s)
- P K Hashim
- Research Institute for Electronic Science, Hokkaido University Kita20, Nishi 10, Kita-ku Sapporo Hokkaido 001-0020 Japan
- Graduate School of Life Science, Hokkaido University Kita 10, Nishi 8, Kita-ku Sapporo Hokkaido 060-0810 Japan
| | - Ashwin T Shaji
- Research Institute for Electronic Science, Hokkaido University Kita20, Nishi 10, Kita-ku Sapporo Hokkaido 001-0020 Japan
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram Kerala 695551 India
| | - Ammathnadu S Amrutha
- Research Institute for Electronic Science, Hokkaido University Kita20, Nishi 10, Kita-ku Sapporo Hokkaido 001-0020 Japan
- Graduate School of Life Science, Hokkaido University Kita 10, Nishi 8, Kita-ku Sapporo Hokkaido 060-0810 Japan
| | - Shifa Ahmad
- Research Institute for Electronic Science, Hokkaido University Kita20, Nishi 10, Kita-ku Sapporo Hokkaido 001-0020 Japan
- Graduate School of Life Science, Hokkaido University Kita 10, Nishi 8, Kita-ku Sapporo Hokkaido 060-0810 Japan
| |
Collapse
|
16
|
Roy S, Pyari G, Bansal H. Theoretical analysis of low-power deep synergistic sono-optogenetic excitation of neurons by co-expressing light-sensitive and mechano-sensitive ion-channels. Commun Biol 2025; 8:379. [PMID: 40050670 PMCID: PMC11885482 DOI: 10.1038/s42003-025-07792-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
The present challenge in neuroscience is to non-invasively exercise low-power and high-fidelity control of neurons situated deep inside the brain. Although, two-photon optogenetic excitation can activate neurons to millimeter depth with sub-cellular specificity and millisecond temporal resolution, it can also cause heating of the targeted tissue. On the other hand, sonogenetics can non-invasively modulate the cellular activity of neurons expressed with mechano-sensitive proteins in deeper areas of the brain with less spatial selectivity. We present a theoretical analysis of a synergistic sono-optogenetic method to overcome these limitations by co-expressing a mechano-sensitive (MscL-I92L) ion-channel with a light-sensitive (CoChR/ChroME2s/ChRmine) ion-channel in hippocampal neurons. It is shown that in the presence of low-amplitude subthreshold ultrasound pulses, the two-photon excitation threshold for neural spiking reduces drastically by 73% with MscL-I92L-CoChR (0.021 mW/µm2), 66% with MscL-I92L-ChroME2s (0.029 mW/µm2), and 64% with MscL-I92L-ChRmine (0.013 mW/µm2) at 5 Hz. It allows deeper excitation of up to 1.2 cm with MscL-I92L-ChRmine combination. The method is useful to design new experiments for low-power deep excitation of neurons and multimodal neuroprosthetic devices and circuits.
Collapse
Affiliation(s)
- Sukhdev Roy
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, India.
| | - Gur Pyari
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, India
| | - Himanshu Bansal
- Department of Physics and Computer Science, Dayalbagh Educational Institute, Agra, India
| |
Collapse
|
17
|
Parekh PK. Illuminating the impact of stress: In vivo approaches to track stress-related neural adaptations. Neurobiol Stress 2025; 35:100712. [PMID: 40191171 PMCID: PMC11970376 DOI: 10.1016/j.ynstr.2025.100712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/16/2024] [Accepted: 02/06/2025] [Indexed: 04/09/2025] Open
Abstract
Stressful experiences can affect both daily life and long-term health outcomes in a variety of ways. Acute challenges may be adaptive, promoting arousal and enhancing memory and cognitive function. Importantly, however, chronic stress dysregulates the body's physiological regulatory mechanisms consisting of complex hormone interactions throughout the peripheral and central nervous systems. This disrupted signaling consequently alters the balance of synapse formation, maturation and pruning, processes which regulate neural communication, plasticity, learning, cognitive flexibility and adaptive behaviors - hallmarks of a healthy, functional brain. The chronically stressed brain state, therefore, is one which may be uniquely vulnerable. To understand the development of this state, how it is sustained and how behavior and neural function are transiently or indelibly impacted by it, we can turn to a number of advanced approaches in animal models which offer unprecedented insights. This has been the aim of my recent work within the field and the goal of my new independent research program. To achieve this, I have employed methods to uncover how key brain circuits integrate information to support motivated behaviors, how stress impacts their ability to perform this process and how best to operationalize behavioral readouts. Here I present an overview of research contributions that I find most meaningful for advancing our understanding of the impact of stress and propose new avenues which will guide my own framework to address the salient outstanding questions within the field.
Collapse
Affiliation(s)
- Puja K. Parekh
- Department of Neuroscience, The University of Texas at Dallas, 860 N. Loop Rd, Richardson, TX, 75080, USA
| |
Collapse
|
18
|
Wang X, Kerckhoffs A, Riexinger J, Cornall M, Langton MJ, Bayley H, Qing Y. ON-OFF nanopores for optical control of transmembrane ionic communication. NATURE NANOTECHNOLOGY 2025; 20:432-440. [PMID: 39838209 PMCID: PMC11919769 DOI: 10.1038/s41565-024-01823-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/09/2024] [Indexed: 01/23/2025]
Abstract
Nanoscale photoswitchable proteins could facilitate precise spatiotemporal control of transmembrane communication and support studies in synthetic biology, neuroscience and bioelectronics. Here, through covalent modification of the α-haemolysin protein pore with arylazopyrazole photoswitches, we produced 'photopores' that transition between iontronic resistor and diode modes in response to irradiation at orthogonal wavelengths. In the diode mode, a low-leak OFF-state nanopore exhibits a reversible increase in unitary conductance of more than 20-fold upon irradiation at 365 nm. A rectification ratio of >5 was achieved with photopores in the diode state by either direct or alternating voltage input. Unlike conventional electronic phototransistors with intensity-dependent photoelectric responses, the photopores regulated current output solely based on the wavelength(s) of monochromatic or dual-wavelength irradiation. Dual-wavelength irradiation at various relative intensities allowed graded adjustment of the photopore conductance. By using these properties, photonic signals encoding text or graphic messages were converted into ionic signals, highlighting the potential applications of photopores as components of smart devices in synthetic biology.
Collapse
Affiliation(s)
- Xingzao Wang
- Department of Chemistry, University of Oxford, Oxford, UK
| | | | | | | | | | - Hagan Bayley
- Department of Chemistry, University of Oxford, Oxford, UK.
| | - Yujia Qing
- Department of Chemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Nam KY, Kim MS, An J, Min S, Lee JH, Park JS, Huh CH, Yun SH, Lee KJ. Human-Centric, Three Dimensional Micro Light-Emitting Diodes for Cosmetic and Medical Phototherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416716. [PMID: 39960366 PMCID: PMC11905057 DOI: 10.1002/advs.202416716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/03/2025] [Indexed: 03/14/2025]
Abstract
Phototherapy based on micro light-emitting diodes (µLEDs) has gained enormous attention in the medical field as a patient-friendly therapeutic method due to its advantages of minimal invasiveness, fewer side effects, and versatile device form factors with high stability in biological environment. Effective cosmetic and medical phototherapy depends on deep light penetration, precise irradiation, and simultaneous multi-site stimulation, facilitated by three-dimensional (3D) optoelectronics specifically designed for complex human matters, defined here as 3D µLEDs. This perspective article aims to present the functionalities and strategies of 3D µLEDs for human-centric phototherapy. This study investigates the effectiveness of phototherapy enabled by three key functionalities such as shape morphing, self-adaptation, and multilayered spatiotemporal mapping of 3D µLEDs. Finally, this article provides future insights of 3D µLEDs for human-centric phototherapy applications.
Collapse
Affiliation(s)
- Ki Yun Nam
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
- School of Electrical Engineering, Graduate School of Semiconductor Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
| | - Min Seo Kim
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
| | - Jaehun An
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
| | - Seongwook Min
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
| | - Jae Hee Lee
- Querrey-Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60 208, USA
| | - Jae Sung Park
- Yonsei Myview Clinic, 301, Sadang-ro, Dongjak-gu, Seoul, 0 7008, Republic of Korea
| | - Chang-Hun Huh
- Department of Dermatology, Seoul National University Bundang Hospital (SNUBH), 173-82, Gumi-ro, Bundang-gu, Seongnam, 13 620, Republic of Korea
| | - Seok Hyun Yun
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, 0 2114, USA
| | - Keon Jae Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
- School of Electrical Engineering, Graduate School of Semiconductor Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34 141, Republic of Korea
| |
Collapse
|
20
|
Govorunova EG, Sineshchekov OA, Li H, Gou Y, Chen H, Yang S, Wang Y, Mitchell S, Palmateer A, Brown LS, St-Pierre F, Xue M, Spudich JL. Blue-shifted ancyromonad channelrhodopsins for multiplex optogenetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639930. [PMID: 40060592 PMCID: PMC11888301 DOI: 10.1101/2025.02.24.639930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Light-gated ion channels from protists (channelrhodopsins or ChRs) are optogenetic tools widely used for controlling neurons and cardiomyocytes. Multiplex optogenetic applications require spectrally separated molecules that must be found in nature, as they are difficult to engineer without disrupting channel function. Scanning numerous sequence databases, we identified three robust naturally blue-shifted ChRs from ancyromonads. They form a separate branch on the phylogenetic tree and contain residue motifs characteristic of anion ChRs (ACRs). However, only two conduct chloride, whereas the close Nutomonas longa homolog (peak absorption at ~440 nm) generates inward cation currents in mammalian cells under physiological conditions, significantly exceeding those by previously known tools. Measurements of transient absorption changes and pH titration of purified Ancyromonas sigmoides ACR (AnsACR) combined with mutant analysis revealed the roles of the residues in the photoactive site. Both ancyromonad ACRs allowed optogenetic silencing of mouse cortical neurons in brain slices. AnsACR expression in the cholinergic neurons enabled photoinhibition of pharyngeal muscle contraction in live worms. AnsACR could be activated by near-infrared two-photon illumination, which is required to control specific neurons in thick tissue. Our results improved the mechanistic understanding of light-gated channel function and expanded the optogenetic toolkit. Impact statement Ancyromonad channelrhodopsins advance our understanding of ionic selectivity and wavelength regulation in light-gated ion channels and also expand the toolkit for all-optical electrophysiology.
Collapse
Affiliation(s)
- Elena G Govorunova
- Center for Membrane Biology, Department of Biochemistry & Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Oleg A Sineshchekov
- Center for Membrane Biology, Department of Biochemistry & Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Hai Li
- Center for Membrane Biology, Department of Biochemistry & Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Yueyang Gou
- Department of Neuroscience, Baylor College of Medicine; Houston, TX 77030, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital; Houston, TX 77030, USA
| | - Hongmei Chen
- Department of Neuroscience, Baylor College of Medicine; Houston, TX 77030, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital; Houston, TX 77030, USA
| | - Shuyuan Yang
- Department of Neuroscience, Baylor College of Medicine; Houston, TX 77030, USA
| | - Yumei Wang
- Center for Membrane Biology, Department of Biochemistry & Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Stephen Mitchell
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph; Guelph, Ontario N1G 2W1, Canada
| | - Alyssa Palmateer
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph; Guelph, Ontario N1G 2W1, Canada
| | - Leonid S Brown
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph; Guelph, Ontario N1G 2W1, Canada
| | - François St-Pierre
- Department of Neuroscience, Baylor College of Medicine; Houston, TX 77030, USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, TX 77005, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 7705, USA
| | - Mingshan Xue
- Department of Neuroscience, Baylor College of Medicine; Houston, TX 77030, USA
- The Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital; Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - John L Spudich
- Center for Membrane Biology, Department of Biochemistry & Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| |
Collapse
|
21
|
Balmer GL, Guha S, Poll S. Engrams across diseases: Different pathologies - unifying mechanisms? Neurobiol Learn Mem 2025; 219:108036. [PMID: 40023216 DOI: 10.1016/j.nlm.2025.108036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Memories are our reservoir of knowledge and thus, are crucial for guiding decisions and defining our self. The physical correlate of a memory in the brain is termed an engram and since decades helps researchers to elucidate the intricate nature of our imprinted experiences and knowledge. Given the importance that memories have for our lives, their impairment can present a tremendous burden. In this review we aim to discuss engram malfunctioning across diseases, covering dementia-associated pathologies, epilepsy, chronic pain and psychiatric disorders. Current neuroscientific tools allow to witness the emergence and fate of engram cells and enable their manipulation. We further suggest that specific mechanisms of mnemonic malfunction can be derived from engram cell readouts. While depicting the way diseases act on the mnemonic component - specifically, on the cellular engram - we emphasize a differentiation between forms of amnesia and hypermnesia. Finally, we highlight commonalities and distinctions of engram impairments on the cellular level across diseases independent of their pathogenic origins and discuss prospective therapeutic measures.
Collapse
Affiliation(s)
- Greta Leonore Balmer
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Shuvrangshu Guha
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany
| | - Stefanie Poll
- University of Bonn, Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research (IEECR), Cellular Neuropathology and Cognition Group, Venusberg-Campus 1/C76, 53127 Bonn, Germany; University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE) Bonn, Germany.
| |
Collapse
|
22
|
Li C, Hu C, Li L, He F, Li X. High-Precision Field- Effect Transistor Biosensor for Analyzing Differential Effects of Anti-Cancer Drugs on Cancerous and Non-Cancerous Cells. BIOSENSORS 2025; 15:125. [PMID: 39997027 PMCID: PMC11852787 DOI: 10.3390/bios15020125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/08/2025] [Accepted: 02/16/2025] [Indexed: 02/26/2025]
Abstract
A high-precision biosensor technique is introduced, offering the capability to independently evaluate the effects of anti-cancer drugs on both cancerous (RAJI) and non-cancerous (WIL2S) cells. By analyzing and fitting current change curves and transfer characteristic curves under two drugs, camptothecin and doxorubicin, this technique quantifies both the magnitude of drug-induced current changes in cells and the rate of drug entry into cells. Flow cytometry was utilized to validate the entry rates of two drugs, camptothecin and doxorubicin, into the cells. The biosensor leverages the exceptional sensitivity of two-dimensional electron gas to detect proximal charge variations at ultralow concentrations, even in fluids with high ionic strength. The findings reveal that anti-cancer drugs have a more pronounced impact on tumor cells, with the effects and interaction speeds differing across normal cells and tumor cells. This innovative approach not only enhances our understanding of the specificity and action mechanisms of anti-cancer drugs but also provides a valuable tool for screening potential tumor anti-cancer drugs and advancing targeted cancer therapies.
Collapse
Affiliation(s)
- Can Li
- Engineering Research Center of TCM Intelligence Health Service, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Can Hu
- School of Computer and Software, Hohai University, Nanjing 211100, China
| | - Ling Li
- Engineering Research Center of TCM Intelligence Health Service, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feng He
- College of Computer and Information Engineering (College of Artificial Intelligence), Nanjing Tech University, Nanjing 211816, China
| | - Xiaofei Li
- Department of Sports Medicine, The First People’s Hospital of Lianyungang, Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang 222599, China
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
23
|
Zhou W, Jia L, Yue L, Hu L. Advances and applications of peripheral optogenetics in animal models. Neuroscience 2025; 567:163-171. [PMID: 39765287 DOI: 10.1016/j.neuroscience.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/27/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
Peripheral optogenetics is an emerging neuromodulation technique that regulates the activity of the nervous system outside the brain through the expression of photosensitive proteins and the application of photic stimulation. This article reviews recent advances in applying optogenetics to the spinal cord and peripheral nerves, offering a comprehensive understanding of the functions and regulatory mechanisms of the peripheral nervous system through the modulation of specific neuronal activities. By showcasing novel opportunities for disease treatment, this technique opens new avenues in psychophysiological research and neural regulation therapy. Despite current challenges, such as operability, effectiveness, and selective neuron targeting, peripheral optogenetics holds significant potential for advancing neuromodulation. Continued research and technological innovations will further expand its role, offering new possibilities for understanding and treating disorders involving the peripheral nervous system.
Collapse
Affiliation(s)
- Wenqian Zhou
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Liping Jia
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Lupeng Yue
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| | - Li Hu
- State Key Laboratory of Cognitive Science and Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
24
|
Yang X, Kubican SE, Yi Z, Tong S. Advances in magnetic nanoparticles for molecular medicine. Chem Commun (Camb) 2025; 61:3093-3108. [PMID: 39846549 PMCID: PMC11756346 DOI: 10.1039/d4cc05167j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Magnetic nanoparticles (MNPs) are highly versatile nanomaterials in nanomedicine, owing to their diverse magnetic properties, which can be tailored through variations in size, shape, composition, and exposure to inductive magnetic fields. Over four decades of research have led to the clinical approval or ongoing trials of several MNP formulations, fueling continued innovation. Beyond traditional applications in drug delivery, imaging, and cancer hyperthermia, MNPs have increasingly advanced into molecular medicine. Under external magnetic fields, MNPs can generate mechano- or thermal stimuli to modulate individual molecules or cells deep within tissue, offering precise, remote control of biological processes at cellular and molecular levels. These unique capabilities have opened new avenues in emerging fields such as genome editing, cell therapies, and neuroscience, underpinned by a growing understanding of nanomagnetism and the molecular mechanisms responding to mechanical and thermal cues. Research on MNPs as a versatile synthetic material capable of engineering control at the cellular and molecular levels holds great promise for advancing the frontiers of molecular medicine, including areas such as genome editing and synthetic biology. This review summarizes recent clinical studies showcasing the classical applications of MNPs and explores their integration into molecular medicine, with the goal of inspiring the development of next-generation MNP-based platforms for disease treatment.
Collapse
Affiliation(s)
- Xiaoyue Yang
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Sarah E Kubican
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Zhongchao Yi
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Sheng Tong
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| |
Collapse
|
25
|
Ban S, Yi H, Park J, Huang Y, Yu KJ, Yeo WH. Advances in Photonic Materials and Integrated Devices for Smart and Digital Healthcare: Bridging the Gap Between Materials and Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416899. [PMID: 39905874 DOI: 10.1002/adma.202416899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/06/2024] [Indexed: 02/06/2025]
Abstract
Recent advances in developing photonic technologies using various materials offer enhanced biosensing, therapeutic intervention, and non-invasive imaging in healthcare. Here, this article summarizes significant technological advancements in materials, photonic devices, and bio-interfaced systems, which demonstrate successful applications for impacting human healthcare via improved therapies, advanced diagnostics, and on-skin health monitoring. The details of required materials, necessary properties, and device configurations are described for next-generation healthcare systems, followed by an explanation of the working principles of light-based therapeutics and diagnostics. Next, this paper shares the recent examples of integrated photonic systems focusing on translation and immediate applications for clinical studies. In addition, the limitations of existing materials and devices and future directions for smart photonic systems are discussed. Collectively, this review article summarizes the recent focus and trends of technological advancements in developing new nanomaterials, light delivery methods, system designs, mechanical structures, material functionalization, and integrated photonic systems to advance human healthcare and digital healthcare.
Collapse
Affiliation(s)
- Seunghyeb Ban
- George W. Woodruff School of Mechanical Engineering, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Hoon Yi
- George W. Woodruff School of Mechanical Engineering, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jaejin Park
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, South Korea
| | - Yunuo Huang
- School of Industrial Design, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Ki Jun Yu
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, South Korea
- The Biotech Center, Pohang University of Science and Technology (POSTECH), Gyeongbuk, 37673, South Korea
- Department of Electrical and Electronic Engineering, YU-Korea Institute of Science and Technology (KIST) Institute, Yonsei University, Seoul, 03722, South Korea
| | - Woon-Hong Yeo
- George W. Woodruff School of Mechanical Engineering, Wearable Intelligent Systems and Healthcare Center at the Institute for Matter and Systems, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
26
|
Morizumi T, Kim K, Li H, Nag P, Dogon T, Sineshchekov OA, Wang Y, Brown LS, Hwang S, Sun H, Bondar AN, Schapiro I, Govorunova EG, Spudich JL, Ernst OP. Structural insights into light-gating of potassium-selective channelrhodopsin. Nat Commun 2025; 16:1283. [PMID: 39900567 PMCID: PMC11790859 DOI: 10.1038/s41467-025-56491-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 01/17/2025] [Indexed: 02/05/2025] Open
Abstract
Structural information on channelrhodopsins' mechanism of light-gated ion conductance is scarce, limiting its engineering as optogenetic tools. Here, we use single-particle cryo-electron microscopy of peptidisc-incorporated protein samples to determine the structures of the slow-cycling mutant C110A of kalium channelrhodopsin 1 from Hyphochytrium catenoides (HcKCR1) in the dark and upon laser flash excitation. Upon photoisomerization of the retinal chromophore, the retinylidene Schiff base NH-bond reorients from the extracellular to the cytoplasmic side. This switch triggers a series of side chain reorientations and merges intramolecular cavities into a transmembrane K+ conduction pathway. Molecular dynamics simulations confirm K+ flux through the illuminated state but not through the resting state. The overall displacement between the closed and the open structure is small, involving mainly side chain rearrangements. Asp105 and Asp116 play a key role in K+ conductance. Structure-guided mutagenesis and patch-clamp analysis reveal the roles of the pathway-forming residues in channel gating and selectivity.
Collapse
Affiliation(s)
- Takefumi Morizumi
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Kyumhyuk Kim
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Hai Li
- Department of Biochemistry & Molecular Biology, Center for Membrane Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Probal Nag
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Physics, Technical University Dortmund, Dortmund, Germany
| | - Tal Dogon
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oleg A Sineshchekov
- Department of Biochemistry & Molecular Biology, Center for Membrane Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Yumei Wang
- Department of Biochemistry & Molecular Biology, Center for Membrane Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Leonid S Brown
- Department of Physics and Biophysics Interdepartmental Group, University of Guelph, Guelph, ON, Canada
| | - Songhwan Hwang
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Han Sun
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Institute of Chemistry, Technical University of Berlin, Berlin, Germany
| | - Ana-Nicoleta Bondar
- Faculty of Physics, University of Bucharest, Măgurele, Romania
- Institute for Computational Biomedicine (INM-9), Forschungszentrum Jülich, Jülich, Germany
| | - Igor Schapiro
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Physics, Technical University Dortmund, Dortmund, Germany
| | - Elena G Govorunova
- Department of Biochemistry & Molecular Biology, Center for Membrane Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - John L Spudich
- Department of Biochemistry & Molecular Biology, Center for Membrane Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA.
| | - Oliver P Ernst
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
27
|
Li Z, Gao Y, Chen X, Xu L, Li Z, Chai R. Study on Recovery Strategy of Hearing Loss & SGN Regeneration Under Physical Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410919. [PMID: 39716878 PMCID: PMC11791950 DOI: 10.1002/advs.202410919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/18/2024] [Indexed: 12/25/2024]
Abstract
The World Health Organization (WHO) reports that by 2050, nearly 2.5 billion people are expected to have some degree of hearing loss (HL) and at least 700 million will need hearing rehabilitation. Therefore, there is an urgent need to develop treatment strategies for HL. At present, the main treatment strategies for HL are hearing aids and cochlear implants (CIs), which cannot achieve a radical cure for HL. Relevant studies have shown that the most fundamental treatment strategy for sensorineural hearing loss (SNHL) is to regenerate hair cells and spiral ganglion neurons (SGNs) through stem cells to repair the structure and function of cochlea. In addition, physical stimulation strategies, such as electricity, light, and magnetism have also been used to promote SGN regeneration. This review systematically introduces the classification, principle and latest progress of the existing hearing treatment strategies and summarizes the advantages and disadvantages of each strategy. The research progress of physical regulation mechanism is discussed in detail. Finally, the problems in HL repair strategies are summarized and the future development direction is prospected, which could provide new ideas and technologies for the optimization of hearing treatment strategies and the research of SGN repair and regeneration through physical regulation.
Collapse
Affiliation(s)
- Zhe Li
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
| | - Yijia Gao
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
| | - Xingyu Chen
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
| | - Lei Xu
- Department of Otolaryngology‐Head and Neck SurgeryShandong Provincial ENT HospitalShandong UniversityJinan250022China
| | - Zhou Li
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
- School of Nanoscience and EngineeringUniversity of Chinese Academy of SciencesBeijing100049China
| | - Renjie Chai
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologySchool of MedicineAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
| |
Collapse
|
28
|
Qin Y, Zhao H, Chang Q, Liu Y, Jing Z, Yu D, Mugo SM, Wang H, Zhang Q. Amylopectin-based Hydrogel Probes for Brain-machine Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416926. [PMID: 39663729 DOI: 10.1002/adma.202416926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Implantable neural probes hold promise for acquiring brain data, modulating neural circuits, and treating various brain disorders. However, traditional implantable probes face significant challenges in practical applications, such as balancing sensitivity with biocompatibility and the difficulties of in situ neural information monitoring and neuromodulation. To address these challenges, this study developed an implantable hydrogel probe capable of recording neural signals, modulating neural circuits, and treating stroke. Amylopectin is integrated into the hydrogels, which can induce reorientation of the poly(3,4-ethylenedioxythiophene) (PEDOT) chain and create compliant interfaces with brain tissues, enhancing both sensitivity and biocompatibility. The hydrogel probe shows the capability of continuously recording deep brain signals for 8 weeks. The hydrogel probe is effectively utilized to study deep brain signals associated with various physiological activities. Neuromodulation and neural signal monitoring are performed directly in the primary motor cortex of rats, enabling control over their limb behaviors through evoked signals. When applied to the primary motor cortex of stroke-affected rats, neuromodulation significantly reduced the brain infarct area, promoted synaptic reorganization, and restored motor functions and balance. This research represents a significant scientific breakthrough in the design of neural probes for brain monitoring, neural circuit modulation, and the development of brain disease therapies.
Collapse
Affiliation(s)
- Yanxia Qin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Hao Zhao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Qi Chang
- Department of Orthopaedics, The 989 Hospital of the People's Liberation Army Joint Service Support Force, Luoyang, 471031, P. R. China
| | - Yan Liu
- Key Laboratory of Bionic Engineering (Ministry of Education), Jilin University, Changchun, 130025, P. R. China
| | - Zhen Jing
- Jilin Provincial Science and Technology Innovation Platform Management Center, Changchun, 130012, P. R. China
| | - Dehai Yu
- Core Facility, The First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, P. R. China
| | - Samuel M Mugo
- Department of Physical Sciences, MacEwan University, Edmonton, ABT5J4S2, Canada
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Qiang Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
29
|
Hedrich R, Gilliham M. Light-activated channelrhodopsins: a revolutionary toolkit for the remote control of plant signalling. THE NEW PHYTOLOGIST 2025; 245:982-988. [PMID: 39632281 DOI: 10.1111/nph.20311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
Channelrhodopsins (CHRs), originating within algae and protists, are membrane-spanning ion channel proteins that are directly activated and/or deactivated by specific wavelengths of light. Since 2005, CHRs have been deployed as genetically encoded optogenetic tools to rapidly advance understanding of neuronal networks. CHRs provide the opportunity to finely tune ion transport across membranes and regulate membrane potential. These are fundamental biochemical signals, which in plants can be translated into physiological and developmental responses such as changes in photosynthesis, growth, turgor, vascular hydraulics, phosphorylation or reactive oxygen species (ROS) status, gene expression, or even cell death. Exploration of CHR family diversity and structure-function engineering has led to the expansion of the CHR optogenetic toolbox, offering unparalleled opportunities to precisely control and understand electrical and secondary messenger signalling in higher plants. In this Tansley Insight, we provide an overview of the recent progress in the application of CHR optogenetics in higher plants and discuss their possible uses in the remote control of plant biology, illuminating a new future domain for plant research enabled through synthetic biology.
Collapse
Affiliation(s)
- Rainer Hedrich
- Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute for Biosciences, Biocenter, Würzburg University, Julius-von-Sachs-Platz 2, D-97082, Würzburg, Germany
| | - Matthew Gilliham
- ARC Centre of Excellence in Plants for Space, School of Agriculture, Food and Wine & Waite Research Institute, University of Adelaide, Urrbrae, SA, 5064, Australia
| |
Collapse
|
30
|
Rahman MA, Hasan MM, Corpas FJ. Leveraging light-gated channelrhodopsins for strengthening plant physiological responses. TRENDS IN PLANT SCIENCE 2025:S1360-1385(25)00010-X. [PMID: 39893118 DOI: 10.1016/j.tplants.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/06/2025] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Strengthening plant physiological traits is crucial for sustainable plant improvement. The underlying molecular mechanisms of rhodopsin-based plant improvement remain largely unknown. However, a recent study by Ding et al. offers some insights by exploring how light-gated channelrhodopsins regulate cytosolic Ca2+ conductance, reactive oxygen species (ROS) signals, and plant defense responses in tobacco.
Collapse
Affiliation(s)
| | - Md Mahadi Hasan
- State Key Laboratory of Herbage Improvement and Grassland Agro-Ecosystems, College of Ecology, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Francisco J Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Stress, Development and Signaling in Plants, Estación Experimental del Zaidín, Profesor Albareda 1, Spanish National Research Council (CSIC), Granada 18008, Spain.
| |
Collapse
|
31
|
Kale MB, Wankhede NL, Bishoyi AK, Ballal S, Kalia R, Arya R, Kumar S, Khalid M, Gulati M, Umare M, Taksande BG, Upaganlawar AB, Umekar MJ, Kopalli SR, Fareed M, Koppula S. Emerging biophysical techniques for probing synaptic transmission in neurodegenerative disorders. Neuroscience 2025; 565:63-79. [PMID: 39608699 DOI: 10.1016/j.neuroscience.2024.11.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Plethora of research has shed light on the critical role of synaptic dysfunction in various neurodegenerative disorders (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Synapses, the fundamental units for neural communication in the brain, are highly vulnerable to pathological conditions and are central to the progression of neurological diseases. The presynaptic terminal, a key component of synapses responsible for neurotransmitter release and synaptic communication, undergoes structural and functional alterations in these disorders. Understanding synaptic transmission abnormalities is crucial for unravelling the pathophysiological mechanisms underlying neurodegeneration. In the quest to probe synaptic transmission in NDDs, emerging biophysical techniques play a pivotal role. These advanced methods offer insights into the structural and functional changes occurring at nerve terminals in conditions like AD, PD, HD & ALS. By investigating synaptic plasticity and alterations in neurotransmitter release dynamics, researchers can uncover valuable information about disease progression and potential therapeutic targets. The review articles highlighted provide a comprehensive overview of how synaptic vulnerability and pathology are shared mechanisms across a spectrum of neurological disorders. In major neurodegenerative diseases, synaptic dysfunction is a common thread linking these conditions. The intricate molecular machinery involved in neurotransmitter release, synaptic vesicle dynamics, and presynaptic protein regulation are key areas of focus for understanding synaptic alterations in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Mohit Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box- 71666, Riyadh 11597, Saudi Arabia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
32
|
Wu D, Sun X, Chen X. Chemo-optogenetic Dimerization Dissects Complex Biological Processes. SMALL METHODS 2025:e2401271. [PMID: 39815164 DOI: 10.1002/smtd.202401271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Light offers superior control in terms of high temporal precision, high spatial precision, and non-invasiveness for the regulation of cellular functions. In recent years, chemical biologists have adopted chemo-optogenetic dimerization approaches, such as photo-triggered chemical inducers of dimerization (pCIDs), as a general tool for spatiotemporal regulation of cellular functions. Traditional chemo-optogenetic dimerization triggers either a single ON or a single OFF of cellular activity. However, more sophisticated approaches are introduced in recent years. These include the ability to turn ON and OFF using different wavelengths of light, tools enabling multi-layer control of cellular activities, and nanobody-tethered photodimerizers. These advancements not only shed light on the study of ubiquitously existing multi-functional proteins but also create new opportunities for investigating complex cellular activity networks.
Collapse
Affiliation(s)
- Donglian Wu
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences, Harbin Institute of Technology (HIT), Harbin, 150001, P. R. China
- Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, P. R. China
| | - Xiaofeng Sun
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences, Harbin Institute of Technology (HIT), Harbin, 150001, P. R. China
- Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, P. R. China
| | - Xi Chen
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences, Harbin Institute of Technology (HIT), Harbin, 150001, P. R. China
- Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, P. R. China
| |
Collapse
|
33
|
Khosravi M, Wieduwilt T, Zeisberger M, Lorenz A, Schmidt MA. Advanced remote focus control in multicore meta-fibers through 3D nanoprinted phase-only holograms. Nat Commun 2025; 16:507. [PMID: 39779667 PMCID: PMC11711281 DOI: 10.1038/s41467-024-55805-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
In this study, we present an unexplored approach for remote focus manipulation using 3D nanoprinted holograms integrated on the end face of multi-core single-mode fibers. This innovative method enables precise focus control within a monolithic metafiber device by allowing light coupled into any of the 37 cores to be precisely focused at predefined locations. Our approach demonstrates significant advances over conventional lenses and offers unique functionalities through computationally designed holograms. This research marks the first successful use of multi-core fibers for remote focus control via 3D nanoprinting, achieving crosstalk-free operation at visible wavelengths. Key findings include strong agreement between design, simulation, and experimental results, highlighting the potential of this technology to improve applications in fields such as biological optics, laser micromachining, telecommunications, and laser surgery. This work opens new avenues for the development of advanced optical systems with superior focus control capabilities.
Collapse
Affiliation(s)
- Mohammadhossein Khosravi
- Leibniz Institute of Photonic Technology, Jena, Germany
- Abbe Center of Photonics and Faculty of Physics, FSU Jena, Jena, Germany
| | | | | | - Adrian Lorenz
- Leibniz Institute of Photonic Technology, Jena, Germany
| | - Markus A Schmidt
- Leibniz Institute of Photonic Technology, Jena, Germany.
- Abbe Center of Photonics and Faculty of Physics, FSU Jena, Jena, Germany.
- Otto Schott Institute of Material Research, FSU Jena, Jena, Germany.
| |
Collapse
|
34
|
Yang Z, Teaney NA, Buttermore ED, Sahin M, Afshar-Saber W. Harnessing the potential of human induced pluripotent stem cells, functional assays and machine learning for neurodevelopmental disorders. Front Neurosci 2025; 18:1524577. [PMID: 39844857 PMCID: PMC11750789 DOI: 10.3389/fnins.2024.1524577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Neurodevelopmental disorders (NDDs) affect 4.7% of the global population and are associated with delays in brain development and a spectrum of impairments that can lead to lifelong disability and even mortality. Identification of biomarkers for accurate diagnosis and medications for effective treatment are lacking, in part due to the historical use of preclinical model systems that do not translate well to the clinic for neurological disorders, such as rodents and heterologous cell lines. Human-induced pluripotent stem cells (hiPSCs) are a promising in vitro system for modeling NDDs, providing opportunities to understand mechanisms driving NDDs in human neurons. Functional assays, including patch clamping, multielectrode array, and imaging-based assays, are popular tools employed with hiPSC disease models for disease investigation. Recent progress in machine learning (ML) algorithms also presents unprecedented opportunities to advance the NDD research process. In this review, we compare two-dimensional and three-dimensional hiPSC formats for disease modeling, discuss the applications of functional assays, and offer insights on incorporating ML into hiPSC-based NDD research and drug screening.
Collapse
Affiliation(s)
- Ziqin Yang
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Nicole A. Teaney
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elizabeth D. Buttermore
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Human Neuron Core, Boston Children’s Hospital, Boston, MA, United States
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Human Neuron Core, Boston Children’s Hospital, Boston, MA, United States
| | - Wardiya Afshar-Saber
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
35
|
Liu Z, Wang L, Wu P, Yuan L. Precision tumor treatment utilizing bacteria: principles and future perspectives. Appl Microbiol Biotechnol 2025; 109:2. [PMID: 39754636 DOI: 10.1007/s00253-024-13378-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/06/2025]
Abstract
Bacteria-based tumor therapy, which releases therapeutic payloads or remodels the tumor's immune-suppressive microenvironment and directly kills tumor cells or initiates an anti-tumor immune response, is recently recognized as a promising strategy. Bacteria could be endowed with the capacities of tumor targeting, tumor cell killing, and anti-tumor immune activating by established gene engineering. Furthermore, the integration of synthetic biology and nanomedicine into these engineered bacteria could further enhance their efficacy and controllability. This comprehensive review systematically elucidates the classification and mechanisms of bacterial gene expression induction systems, as well as strategies for constructing bacterial-nanomaterial nanobiohybrids. The review concludes by highlighting the challenges associated with quality control and regulation of bacteria-based tumor therapy while also providing insights into the future prospects of this therapeutic technology. KEY POINTS: • A comprehensive overview of the current status of research on bacteria-based tumor therapy. • The classification and mechanisms of bacterial gene expression induction systems are summarized. • The challenges and perspectives in clinical translation.
Collapse
Affiliation(s)
- Zhaoyou Liu
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Lantian Wang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Pengying Wu
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Lijun Yuan
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Air Force Medical University, No.569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
36
|
Huang J, Fussenegger M. Programming mammalian cell behaviors by physical cues. Trends Biotechnol 2025; 43:16-42. [PMID: 39179464 DOI: 10.1016/j.tibtech.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/26/2024]
Abstract
In recent decades, the field of synthetic biology has witnessed remarkable progress, driving advances in both research and practical applications. One pivotal area of development involves the design of transgene switches capable of precisely regulating specified outputs and controlling cell behaviors in response to physical cues, which encompass light, magnetic fields, temperature, mechanical forces, ultrasound, and electricity. In this review, we delve into the cutting-edge progress made in the field of physically controlled protein expression in engineered mammalian cells, exploring the diverse genetic tools and synthetic strategies available for engineering targeting cells to sense these physical cues and generate the desired outputs accordingly. We discuss the precision and efficiency limitations inherent in these tools, while also highlighting their immense potential for therapeutic applications.
Collapse
Affiliation(s)
- Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland; Faculty of Science, University of Basel, Klingelbergstrasse 48, CH-4056 Basel, Switzerland.
| |
Collapse
|
37
|
Wang X, Yu Y, Wang Q. Modeling the modulation of beta oscillations in the basal ganglia by dual-target optogenetic stimulation. FUNDAMENTAL RESEARCH 2025; 5:82-92. [PMID: 40166095 PMCID: PMC11955041 DOI: 10.1016/j.fmre.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/11/2023] [Accepted: 01/15/2024] [Indexed: 04/02/2025] Open
Abstract
Optogenetic techniques provide precise control over the activity of specific neurons within the nucleus, offering more accurate regulatory effects compared to deep brain stimulation. The heterogeneity of the globus pallidus externa (GPe) has garnered wide attention, wherein significant differences in pathological changes emphasize its potential as a stimulation target with distinct mechanisms. A basal ganglia-thalamus (BG-Th) network model incorporating heterogeneous GPe is developed to explore potential optogenetic stimulation targets for treating Parkinson's disease (PD). Initially, the modulation mechanisms of single-target optogenetic stimulation on the abnormal rhythmic oscillations of BG nuclei are examined. Excitation of D1 medium spine neuron (MSN), calcium-binding protein parvalbumin (PV) GPe, and inhibition of globus pallidus interna (GPi) can effectively suppress synchronous bursting activity in GPi, while excitation of GPi promotes high-frequency discharge to disrupt beta oscillations. Furthermore, dual-target optogenetic stimulation strategies are devised to reduce energy consumption. Results show that targets with similar mechanisms exhibit additive effects, whereas targets with opposing mechanisms lead to cancellation. The underlying effective mechanisms of dual-target strategies are: enhancing the inhibitory input to GPi thus inhibiting the activity of GPi, or disrupting beta oscillations by restoring high-frequency discharges in GPi. The strategy composed of exciting D1 MSN and inhibiting GPi requires the minimum total light intensity among single-target and dual-target strategies in our simulation. Furthermore, simultaneously enhancing PV GPe and inhibiting D2 MSN achieves the greatest reduction in total energy consumption (40.8% reduction), compared to only enhancing PV GPe. The findings unveil effective circuit mechanisms of optogenetic stimulation and provide novel insights for designing precise regulatory strategies for PD.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Dynamics and Control, Beihang University, Beijing 100191, China
| | - Ying Yu
- Department of Dynamics and Control, Beihang University, Beijing 100191, China
| | - Qingyun Wang
- Department of Dynamics and Control, Beihang University, Beijing 100191, China
- School of Mathematics and Statistics, Ningxia University, Yinchuan 750021, China
- Ningxia Basic Science Research Center of Mathematics, Yinchuan 750021, China
| |
Collapse
|
38
|
Hmidi D, Muraya F, Fizames C, Véry A, Roelfsema MRG. Potassium extrusion by plant cells: evolution from an emergency valve to a driver of long-distance transport. THE NEW PHYTOLOGIST 2025; 245:69-87. [PMID: 39462778 PMCID: PMC11617655 DOI: 10.1111/nph.20207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 08/15/2024] [Indexed: 10/29/2024]
Abstract
The ability to accumulate nutrients is a hallmark for living creatures and plants evolved highly effective nutrient transport systems, especially for the uptake of potassium (K+). However, plants also developed mechanisms that enable the rapid extrusion of K+ in combination with anions. The combined release of K+ and anions is probably an ancient extrusion system, as it is found in the Characeae that are closely related to land plants. We postulate that the ion extrusion mechanisms have developed as an emergency valve, which enabled plant cells to rapidly reduce their turgor, and prevent them from bursting. Later in evolution, seed plants adapted this system for various responses, such as the closure of stomata, long-distance stress waves, dropping of leaves by pulvini, and loading of xylem vessels. We discuss the molecular nature of the transport proteins that are involved in ion extrusion-based functions of plants and describe the functions that they obtained during evolution.
Collapse
Affiliation(s)
- Dorsaf Hmidi
- Institut des Sciences des Plantes de Montpellier, Univ Montpellier, CNRS, INRAE, Institut Agro, Campus SupAgro‐INRAE34060Montpellier Cedex 2France
| | - Florence Muraya
- Molecular Plant Physiology and Biophysics, Julius‐von‐Sachs Institute for Biosciences, BiocenterWürzburg UniversityJulius‐von‐Sachs‐Platz 2D‐97082WürzburgGermany
| | - Cécile Fizames
- Institut des Sciences des Plantes de Montpellier, Univ Montpellier, CNRS, INRAE, Institut Agro, Campus SupAgro‐INRAE34060Montpellier Cedex 2France
| | - Anne‐Aliénor Véry
- Institut des Sciences des Plantes de Montpellier, Univ Montpellier, CNRS, INRAE, Institut Agro, Campus SupAgro‐INRAE34060Montpellier Cedex 2France
| | - M. Rob G. Roelfsema
- Molecular Plant Physiology and Biophysics, Julius‐von‐Sachs Institute for Biosciences, BiocenterWürzburg UniversityJulius‐von‐Sachs‐Platz 2D‐97082WürzburgGermany
| |
Collapse
|
39
|
Shkarina K, Broz P. Targeted Activation of Programmed Cell Death Pathways by Optogenetics. Methods Mol Biol 2025; 2840:57-74. [PMID: 39724344 DOI: 10.1007/978-1-0716-4047-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Regulated cell death is an important biological process by which an organism removes unwanted, malignant, or infected cells. Although it has become clear that different forms of regulated cell death exist, it remains difficult to compare their consequences at the cellular and tissue level as they are induced by different stimuli and proceed with different kinetics. Moreover, it was so far difficult to target and induce cell death in selected cells within cell populations or complex tissues without affecting its neighbors. To overcome these obstacles researchers developed novel synthetic biology tools that use chemical or optogenetic approaches to specifically induce PCD in selected cells. Here, we describe a protocol for optogenetic activation of three major types of regulated cell death: apoptosis, necroptosis, and pyroptosis, using light-induced forced oligomerization of their major effector proteins (caspases or kinases).
Collapse
Affiliation(s)
- Kateryna Shkarina
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany.
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
40
|
Chen X, Feng Y, Zhang P, Ni Z, Xue Y, Liu J. Hydrogel Fibers-Based Biointerfacing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413476. [PMID: 39578344 DOI: 10.1002/adma.202413476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/12/2024] [Indexed: 11/24/2024]
Abstract
The unique 1D structure of fibers offers intriguing attributes, including a high length-to-diameter ratio, miniatured size, light-weight, and flexibility, making them suitable for various biomedical applications, such as health monitoring, disease treatment, and minimally invasive surgeries. However, traditional fiber devices, typically composed of rigid, dry, and non-living materials, are intrinsically different from the soft, wet, and living essence of biological tissues, thereby posing grand challenges for long-term, reliable, and seamless interfacing with biological systems. Hydrogel fibers have recently emerged as a promising candidate, in light of their similarity to biological tissues in mechanical, chemical and biological aspects, as well as distinct fiber geometry. In this review, a comprehensive overview of recent progress in hydrogel fibers-based biointerfacing technology is provided. It thoroughly summarizes the manufacturing strategy and functional design, especially for hydrogel fibers with distinct optical and electron conductive performance, as well as responsiveness to triggers including thermal, magnetic field and ultrasonic wave, etc. Such unique attributes enable various biomedical applications, which are also examined in detail. Future challenges and potential directions, including biosafety, long-term reliability, sterilization, multi-modalities integration and intelligent therapeutic systems, are raised. This review will serve as a valuable resource for further advancement and implementation as next-generation biointerfacing technology.
Collapse
Affiliation(s)
- Xingmei Chen
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yinghui Feng
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Pei Zhang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhipeng Ni
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yu Xue
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ji Liu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
41
|
Ye X, Qin K, Fernie AR, Zhang Y. Prospects for synthetic biology in 21 st Century agriculture. J Genet Genomics 2024:S1673-8527(24)00369-2. [PMID: 39742963 DOI: 10.1016/j.jgg.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
Plant synthetic biology has emerged as a transformative field in agriculture, offering innovative solutions to enhance food security, provide resilience to climate change, and transition to sustainable farming practices. By integrating advanced genetic tools, computational modeling, and systems biology, researchers can precisely modify plant genomes to enhance traits such as yield, stress tolerance, and nutrient use efficiency. The ability to design plants with specific characteristics tailored to diverse environmental conditions and agricultural needs holds great potential to address global food security challenges. Here, we highlight recent advancements and applications of plant synthetic biology in agriculture, focusing on key areas such as photosynthetic efficiency, nitrogen fixation, drought tolerance, pathogen resistance, nutrient use efficiency, biofortification, climate resilience, microbiology engineering, synthetic plant genomes, and the integration of artificial intelligence (AI) with synthetic biology. These innovations aim to maximize resource use efficiency, reduce reliance on external inputs, and mitigate environmental impacts associated with conventional agricultural practices. Despite challenges related to regulatory approval and public acceptance, the integration of synthetic biology in agriculture holds immense promise for creating more resilient and sustainable agricultural systems, contributing to global food security and environmental sustainability. Rigorous multi-field testing of these approaches will undoubtedly be required to ensure reproducibility.
Collapse
Affiliation(s)
- Xingyan Ye
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kezhen Qin
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Alisdair R Fernie
- Max-Planck-Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam-Golm, Germany.
| | - Youjun Zhang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
42
|
Jafarbeglou F, Dunlop MJ. Red Light Responsive Cre Recombinase for Bacterial Optogenetics. ACS Synth Biol 2024; 13:3991-4001. [PMID: 39558834 DOI: 10.1021/acssynbio.4c00388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Optogenetic tools have been used in a wide range of microbial engineering applications that benefit from the tunable, spatiotemporal control that light affords. However, the majority of current optogenetic constructs for bacteria respond to blue light, limiting the potential for multichromatic control. In addition, other wavelengths offer potential benefits over blue light, including improved penetration of dense cultures and reduced potential for toxicity. In this study, we introduce OptoCre-REDMAP, a red light inducible Cre recombinase system in Escherichia coli. This system harnesses the plant photoreceptors PhyA and FHY1 and a split version of Cre recombinase to achieve precise control over gene expression and DNA excision. We optimized the design by modifying the start codon of Cre and characterized the impact of different levels of induction to find conditions that produced minimal basal expression in the dark and induced full activation within 4 h of red light exposure. We characterized the system's sensitivity to ambient light, red light intensity, and exposure time, finding OptoCre-REDMAP to be reliable and flexible across a range of conditions. In coculture experiments with OptoCre-REDMAP and the blue light responsive OptoCre-VVD, we found that the systems responded orthogonally to red and blue light inputs. Direct comparisons between red and blue light induction with OptoCre-REDMAP and OptoCre-VVD demonstrated the superior penetration properties of red light. OptoCre-REDMAP's robust and selective response to red light makes it suitable for advanced synthetic biology applications, particularly those requiring precise multichromatic control.
Collapse
Affiliation(s)
- Fereshteh Jafarbeglou
- Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Biological Design Center, Boston University, Boston, Massachusetts 02215, United States
| | - Mary J Dunlop
- Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
- Biological Design Center, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
43
|
Vogt A, Paulat R, Parthier D, Just V, Szczepek M, Scheerer P, Xu Q, Möglich A, Schmitz D, Rost BR, Wenger N. Simultaneous spectral illumination of microplates for high-throughput optogenetics and photobiology. Biol Chem 2024; 405:751-763. [PMID: 39303162 DOI: 10.1515/hsz-2023-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
The biophysical characterization and engineering of optogenetic tools and photobiological systems has been hampered by the lack of efficient methods for spectral illumination of microplates for high-throughput analysis of action spectra. Current methods to determine action spectra only allow the sequential spectral illumination of individual wells. Here we present the open-source RainbowCap-system, which combines LEDs and optical filters in a standard 96-well microplate format for simultaneous and spectrally defined illumination. The RainbowCap provides equal photon flux for each wavelength, with the output of the LEDs narrowed by optical bandpass filters. We validated the RainbowCap for photoactivatable G protein-coupled receptors (opto-GPCRs) and enzymes for the control of intracellular downstream signaling. The simultaneous, spectrally defined illumination provides minimal interruption during time-series measurements, while resolving 10 nm differences in the action spectra of optogenetic proteins under identical experimental conditions. The RainbowCap is also suitable for studying the spectral dependence of light-regulated gene expression in bacteria, which requires illumination over several hours. In summary, the RainbowCap provides high-throughput spectral illumination of microplates, while its modular, customizable design allows easy adaptation to a wide range of optogenetic and photobiological applications.
Collapse
Affiliation(s)
- Arend Vogt
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Raik Paulat
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Faculty of Energy and Information, HTW-Berlin University for Applied Sciences, D-10318 Berlin, Germany
| | - Daniel Parthier
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Verena Just
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Faculty of Energy and Information, HTW-Berlin University for Applied Sciences, D-10318 Berlin, Germany
| | - Michal Szczepek
- Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Patrick Scheerer
- Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Qianzhao Xu
- Department of Biochemistry, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Andreas Möglich
- Department of Biochemistry, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Dietmar Schmitz
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- 638588 German Center for Neurodegenerative Diseases (DZNE) , D-10117 Berlin, Germany
| | - Benjamin R Rost
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- 638588 German Center for Neurodegenerative Diseases (DZNE) , D-10117 Berlin, Germany
| | - Nikolaus Wenger
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| |
Collapse
|
44
|
Zheng R, Xue Z, You M. Optogenetic Tools for Regulating RNA Metabolism and Functions. Chembiochem 2024; 25:e202400615. [PMID: 39316432 DOI: 10.1002/cbic.202400615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 09/25/2024]
Abstract
RNA molecules play a vital role in linking genetic information with various cellular processes. In recent years, a variety of optogenetic tools have been engineered for regulating cellular RNA metabolism and functions. These highly desirable tools can offer non-intrusive control with spatial precision, remote operation, and biocompatibility. Here, we would like to review these currently available approaches that can regulate RNAs with light: from non-genetically encodable chemically modified oligonucleotides to genetically encoded RNA aptamers that recognize photosensitive small-molecule or protein ligands. Some key applications of these optogenetic tools will also be highlighted to illustrate how they have been used for regulating all aspects of the RNA life cycle: from RNA synthesis, maturation, modification, and translation to their degradation, localization, and phase separation control. Some current challenges and potential practical utilizations of these RNA optogenetic tools will also be discussed.
Collapse
Affiliation(s)
- Ru Zheng
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Zhaolin Xue
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Mingxu You
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
45
|
A brain circuit that links hunger signals with the jaw movements of eating. Nature 2024:10.1038/d41586-024-03913-1. [PMID: 39633116 DOI: 10.1038/d41586-024-03913-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
|
46
|
Takaramoto S, Fainsod S, Nagata T, Rozenberg A, Béjà O, Inoue K. HulaCCR1, a pump-like cation channelrhodopsin discovered in a lake microbiome. J Mol Biol 2024; 436:168844. [PMID: 39476949 DOI: 10.1016/j.jmb.2024.168844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/27/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Channelrhodopsins are light-gated ion channels consisting of seven transmembrane helices and a retinal chromophore, which are used as popular optogenetic tools for modulating neuronal activity. Cation channelrhodopsins (CCRs), first recognized as the photoreceptors in the chlorophyte Chlamydomonas reinhardtii, have since been identified in diverse species of green algae, as well in other unicellular eukaryotes. The CCRs from non-chlorophyte species are commonly referred to as bacteriorhodopsin-like cation channelrhodopsins, or BCCRs, as most of them feature the three characteristic amino acid residues of the "DTD motif" in the third transmembrane helix (TM3 or helix C) matching the canonical DTD motif of the well-studied archaeal light-driven proton pump bacteriorhodopsin. Here, we report characterization of HulaCCR1, a novel BCCR identified through metatranscriptomic analysis of a unicellular eukaryotic community in Lake Hula, Israel. Interestingly, HulaCCR1 has an ETD motif in which the first residue of the canonical motif is substituted for glutamate. Electrophysiological measurements of the wild-type and a mutant with a DTD motif of HulaCCR1 suggest the critical role of the first glutamate in spectral tuning and channel gating. Additionally, HulaCCR1 exhibits long extensions at the N- and C-termini. Photocurrents recorded from a truncated variant without the signal peptide predicted at the N-terminus were diminished, and membrane localization of the truncated variant significantly decreased, indicating that the signal peptide is important for membrane trafficking of HulaCCR1. These characteristics of HulaCCR1 would be related to a new biological significance in the original unidentified species, distinct from those known for other BCCRs.
Collapse
Affiliation(s)
- Shunki Takaramoto
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Chiba 277-8581, Japan
| | - Shai Fainsod
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Takashi Nagata
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Chiba 277-8581, Japan
| | - Andrey Rozenberg
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Oded Béjà
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; The Nancy and Stephen Grand Technion Energy Program (GTEP), Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Keiichi Inoue
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Chiba 277-8581, Japan.
| |
Collapse
|
47
|
Kim J, Gilbert E, Arndt K, Huang H, Oleniacz P, Jiang S, Kimbrough I, Sontheimer H, English DF, Jia X. Multifunctional Tetrode-like Drug delivery, Optical stimulation, and Electrophysiology (Tetro-DOpE) probes. Biosens Bioelectron 2024; 265:116696. [PMID: 39208508 PMCID: PMC11475332 DOI: 10.1016/j.bios.2024.116696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Having reliable tools for recording and manipulating circuit activity are essential to understand the complex patterns of neural dynamics that underlie brain function. We present Tetro-DOpE (Tetrode-like Drug delivery, Optical stimulation, and Electrophysiology) probes that can simultaneously record and manipulate neural activity in behaving rodents. We fabricated thin multifunctional fibers (<50 μm) using the scalable convergence thermal drawing process. Then, the thin fibers are bundled, similar to tetrode fabrication, to produce Tetro-DOpE probes. We demonstrated the multifunctionality (i.e., electrophysiology, optical stimulation, and drug delivery) of our probe in head-fixed behaving mice. Furthermore, we assembled a six-shank probe mounted on a microdrive which enabled stable recordings of over months when chronically implanted in freely behaving mice. These in vivo experiments demonstrate the potential of customizable, low cost, and accessible multifunctional Tetro-DOpE probes for investigation of neural circuitry in behaving animals.
Collapse
Affiliation(s)
- Jongwoon Kim
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Earl Gilbert
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - Kaiser Arndt
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - Hengji Huang
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Patrycja Oleniacz
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Shan Jiang
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Ian Kimbrough
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Harald Sontheimer
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | | | - Xiaoting Jia
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA; School of Neuroscience, Virginia Tech, Blacksburg, VA, USA; Department of Materials Science and Engineering, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
48
|
Tan HL, Yin L, Tan Y, Ivanov J, Plucinska K, Ilanges A, Herb BR, Wang P, Kosse C, Cohen P, Lin D, Friedman JM. Leptin-activated hypothalamic BNC2 neurons acutely suppress food intake. Nature 2024; 636:198-205. [PMID: 39478220 PMCID: PMC11618066 DOI: 10.1038/s41586-024-08108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 09/24/2024] [Indexed: 11/04/2024]
Abstract
Leptin is an adipose tissue hormone that maintains homeostatic control of adipose tissue mass by regulating the activity of specific neural populations controlling appetite and metabolism1. Leptin regulates food intake by inhibiting orexigenic agouti-related protein (AGRP) neurons and activating anorexigenic pro-opiomelanocortin (POMC) neurons2. However, whereas AGRP neurons regulate food intake on a rapid time scale, acute activation of POMC neurons has only a minimal effect3-5. This has raised the possibility that there is a heretofore unidentified leptin-regulated neural population that rapidly suppresses appetite. Here we report the discovery of a new population of leptin-target neurons expressing basonuclin 2 (Bnc2) in the arcuate nucleus that acutely suppress appetite by directly inhibiting AGRP neurons. Opposite to the effect of AGRP activation, BNC2 neuronal activation elicited a place preference indicative of positive valence in hungry but not fed mice. The activity of BNC2 neurons is modulated by leptin, sensory food cues and nutritional status. Finally, deleting leptin receptors in BNC2 neurons caused marked hyperphagia and obesity, similar to that observed in a leptin receptor knockout in AGRP neurons. These data indicate that BNC2-expressing neurons are a key component of the neural circuit that maintains energy balance, thus filling an important gap in our understanding of the regulation of food intake and leptin action.
Collapse
Affiliation(s)
- Han L Tan
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Luping Yin
- Department of Psychiatry, Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA
| | - Yuqi Tan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessica Ivanov
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Kaja Plucinska
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Anoj Ilanges
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Brian R Herb
- Department of Pharmacology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Putianqi Wang
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Christin Kosse
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Dayu Lin
- Department of Psychiatry, Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
49
|
Sistemich L, Ebbinghaus S. Heat application in live cell imaging. FEBS Open Bio 2024; 14:1940-1954. [PMID: 39489617 PMCID: PMC11609584 DOI: 10.1002/2211-5463.13912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Thermal heating of biological samples allows to reversibly manipulate cellular processes with high temporal and spatial resolution. Manifold heating techniques in combination with live-cell imaging were developed, commonly tailored to customized applications. They include Peltier elements and microfluidics for homogenous sample heating as well as infrared lasers and radiation absorption by nanostructures for spot heating. A prerequisite of all techniques is that the induced temperature changes are measured precisely which can be the main challenge considering subcellular structures or multicellular organisms as target regions. This article discusses heating and temperature sensing techniques for live-cell imaging, leading to future applications in cell biology.
Collapse
Affiliation(s)
- Linda Sistemich
- Chair of Biophysical ChemistryRuhr‐University BochumGermany
- Research Center Chemical Sciences and Sustainability, Research Alliance RuhrBochumGermany
| | - Simon Ebbinghaus
- Chair of Biophysical ChemistryRuhr‐University BochumGermany
- Research Center Chemical Sciences and Sustainability, Research Alliance RuhrBochumGermany
| |
Collapse
|
50
|
Zhu M, Fang Y, Sun Y, Li S, Yu J, Xiong B, Wen C, Zhou B, Huang B, Yin H, Xu H. Sonogenetics in the Treatment of Chronic Diseases: A New Method for Cell Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407373. [PMID: 39488795 DOI: 10.1002/advs.202407373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Sonogenetics is an innovative technology that integrates ultrasound with genetic editing to precisely modulate cellular activities in a non-invasive manner. This method entails introducing and activating mechanosensitive channels on the cell membrane of specific cells using gene delivery vectors. When exposed to ultrasound, these channels can be manipulated to open or close, thereby impacting cellular functions. Sonogenetics is currently being used extensively in the treatment of various chronic diseases, including Parkinson's disease, vision restoration, and cancer therapy. This paper provides a comprehensive review of key components of sonogenetics and focuses on evaluating its prospects and potential challenges in the treatment of chronic disease.
Collapse
Affiliation(s)
- Mingrui Zhu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yan Fang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yikang Sun
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Shaoyue Li
- Department of Medical Ultrasound, Center of Minimally Invasive Treatment for Tumor, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Clinical Research Center for Interventional Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jifeng Yu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bing Xiong
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Congjian Wen
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Boyang Zhou
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Bin Huang
- Zhejiang Hospital, Hangzhou, 310013, P. R. China
| | - Haohao Yin
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Huixiong Xu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|