1
|
Piantadosi PT, Princz-Lebel O, Skirzewski M, Dumont JR, Palmer D, Memar S, Saksida LM, Prado VF, Prado MAM, Bussey TJ, Holmes A. Integrating optical neuroscience tools into touchscreen operant systems. Nat Protoc 2025:10.1038/s41596-025-01143-x. [PMID: 40410621 DOI: 10.1038/s41596-025-01143-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/18/2024] [Indexed: 05/25/2025]
Abstract
Unlocking the neural regulation of complex behavior is a foundational goal of brain science. Touchscreen-based assessments of behavior have been used extensively in the pursuit of this goal, with traditional pharmacological and neurochemical approaches being employed to provide key insights into underlying neural systems. So far, optically based approaches to measure and manipulate neural function, which have begun to revolutionize our understanding of relatively simple behaviors, have been less widely adopted for more complex cognitive functions of the type assessed with touchscreen-based behavioral tasks. Here we provide guidance and procedural descriptions to enable researchers to integrate optically based manipulation and measurement techniques into their touchscreen experimental systems. We focus primarily on three techniques, optogenetic manipulation, fiber photometry and microendoscopic imaging, describing experimental design adjustments that we have found to be critical to the successful integration of these approaches with extant touchscreen behavior pipelines. These include factors related to surgical procedures and timing, alterations to touchscreen operant environments and approaches to synchronizing light delivery and task design. A detailed protocol is included for each of the three techniques, covering their use from implementation through data analysis. The procedures in this protocol can be conducted in as short a time as a few days or over the course of weeks or months.
Collapse
Affiliation(s)
- Patrick T Piantadosi
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Oren Princz-Lebel
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Miguel Skirzewski
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Julie R Dumont
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Daniel Palmer
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Sara Memar
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Lisa M Saksida
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Vania F Prado
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Tim J Bussey
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| |
Collapse
|
2
|
Brumage L, Best S, Hippe DS, Grunblatt E, Chanana P, Wu F, Lee MC, Ying Z, Ibrahim A, Chung JH, Vigil A, Fatherree J, Beronja S, Paddison P, Sullivan L, Nabet B, MacPherson D. In vivo functional screens reveal KEAP1 loss as a driver of chemoresistance in small cell lung cancer. SCIENCE ADVANCES 2025; 11:eadq7084. [PMID: 40267200 PMCID: PMC12017300 DOI: 10.1126/sciadv.adq7084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
Exquisitely chemosensitive initially, small cell lung cancer (SCLC) exhibits dismal outcomes owing to rapid transition to chemoresistance. Elucidating the genetic underpinnings has been challenging owing to limitations with cellular models. As SCLC patient-derived xenograft (PDX) models mimic therapeutic responses, we perform genetic screens in chemosensitive PDX models to identify drivers of chemoresistance. cDNA overexpression screens identify MYC, MYCN, and MYCL, while CRISPR deletion screens identify KEAP1 loss as driving chemoresistance. Deletion of KEAP1 switched a chemosensitive SCLC PDX model to become chemoresistant and resulted in sensitivity to inhibition of glutamine metabolism. Data from the IMpower133 clinical trial revealed ~6% of patients with extensive-stage SCLC exhibit KEAP1 genetic alterations, with activation of a KEAP1/NRF2 transcriptional signature associated with reduced survival upon chemotherapy treatment. While roles for KEAP1/NRF2 have been unappreciated in SCLC, our genetic screens revealed KEAP1 loss as a driver of chemoresistance, while patient genomic analyses demonstrate clinical importance.
Collapse
Affiliation(s)
- Lauren Brumage
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Scott Best
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Eli Grunblatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Pritha Chanana
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Zhe Ying
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ali Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anna Vigil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jackson Fatherree
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patrick Paddison
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lucas Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
3
|
Franks NP, Wisden W. Reply to: A curious concept of CNS clearance. Nat Neurosci 2025; 28:734-736. [PMID: 40069362 DOI: 10.1038/s41593-025-01898-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/28/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute, Imperial College London, London, UK.
| |
Collapse
|
4
|
Hou X, Liu L, Sun L. Precise modulation of cell activity using sono-responsive nano-transducers. Biomaterials 2025; 314:122857. [PMID: 39357155 DOI: 10.1016/j.biomaterials.2024.122857] [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: 07/16/2024] [Revised: 09/09/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Ultrasound, as a form of mechanical energy, possesses a distinctive ability to deeply penetrate tissues, allowing for non-invasive manipulation of cellular activities. Utilizing nanomaterials in conjunction with ultrasound has enabled simple, efficient, spatiotemporally controllable, and minimally invasive regulation of cellular activities with ultrasound-generated electric, optical, acoustic, or chemical stimuli at the localized nanomaterials interface. This technology allows for precise and localized regulation of cellular activities, which is essential for studying and understanding complex biological processes, and also provides new opportunities for research, diagnostics, and therapeutics in the fields of biology and medicine. In this article, we review the state-of-the-art and ongoing developments in nanomaterials-enabled ultrasound cellular modulation, highlighting potential applications and advancements achieved through the integration of sono-responsive nanomaterials with ultrasound.
Collapse
Affiliation(s)
- Xuandi Hou
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, 999077, PR China
| | - Langzhou Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, 999077, PR China
| | - Lei Sun
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, 999077, PR China.
| |
Collapse
|
5
|
Fry BR, Russell N, Fex V, Mo B, Pence N, Beatty JA, Manfredsson FP, Toth BA, Burgess CR, Gershman S, Johnson AW. Devaluing memories of reward: a case for dopamine. Commun Biol 2025; 8:161. [PMID: 39900665 PMCID: PMC11790953 DOI: 10.1038/s42003-024-07440-7] [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: 03/06/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Midbrain dopamine cells encode differences in predictive and expected value to support learning through reward prediction error. Recent findings have questioned whether reward prediction error can fully account for dopamine function and suggest a more complex role for dopamine in encoding detailed features of the reward environment. In this series of studies, we describe a novel role for dopamine in devaluing sensory features of reward. Mesencephalic dopamine cells activated during a mediated devaluation phase were later chemogenetically reactivated. This retrieval of the devalued reward memory elicited a reduction in the hedonic evaluation of sucrose reward. Through optogenetic and chemogenetic manipulations, we confirm dopamine cells are both sufficient and necessary for mediated devaluation, and retrieval of these memories reflected dopamine release in the nucleus accumbens. Consistent with our computational modeling data, our findings indicate a critical role for dopamine in encoding predictive representations of the sensory features of reinforcement. Overall, we elucidate a novel role for dopamine function in mediated devaluation and illuminate a more elaborate framework through which dopamine encodes reinforcement signals.
Collapse
Affiliation(s)
- Benjamin R Fry
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, USA
| | - Nicolette Russell
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Victoria Fex
- Lyman Briggs College, Michigan State University, East Lansing, MI, USA
| | - Bing Mo
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Nathan Pence
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Joseph A Beatty
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Fredric P Manfredsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Brandon A Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Samuel Gershman
- Department of Psychology, Harvard University, Cambridge, MA, USA
| | - Alexander W Johnson
- Department of Psychology, Michigan State University, East Lansing, MI, USA.
- Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
6
|
Doi Y, Asaka M, Born RT, Yanagihara D, Uchida N. A novel behavioral paradigm using mice to study predictive postural control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.01.601478. [PMID: 39005260 PMCID: PMC11244922 DOI: 10.1101/2024.07.01.601478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Postural control circuitry performs the essential function of maintaining balance and body position in response to perturbations that are either self-generated (e.g. reaching to pick up an object) or externally delivered (e.g. being pushed by another person). Human studies have shown that anticipation of predictable postural disturbances can modulate such responses. This indicates that postural control could involve higher-level neural structures associated with predictive functions, rather than being purely reactive. However, the underlying neural circuitry remains largely unknown. To enable studies of predictive postural control circuits, we developed a novel experimental paradigm for mice. In this paradigm, modeled after human studies, a dynamic platform generated reproducible translational perturbations. While mice stood bipedally atop a perch to receive water rewards, they experienced backward translations that were either unpredictable or preceded by an auditory cue. To validate the paradigm, we investigated the effect of the auditory cue on postural responses to perturbations across multiple days in three mice. These preliminary results serve to validate a new postural control experimental paradigm, opening the door to the types of neural recordings and circuit manipulations that are currently possible in mice.
Collapse
Affiliation(s)
- Yurika Doi
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Meiko Asaka
- Cognition and Behavior Joint Research Laboratory, RIKEN center for Brain Science, Wako, Japan
| | - Richard T. Born
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Dai Yanagihara
- Cognition and Behavior Joint Research Laboratory, RIKEN center for Brain Science, Wako, Japan
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Center for Brain Science, Harvard University, Cambridge, MA, USA
| |
Collapse
|
7
|
Villamarin-Ortiz A, Reiche CF, Federer F, Clark AM, Rolston JD, Soto-Sánchez C, Fernandez E, Blair S, Angelucci A. Cortical Response to Acute Implantation of the Utah Optrode Array in Macaque Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632843. [PMID: 39868287 PMCID: PMC11761502 DOI: 10.1101/2025.01.13.632843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Optogenetics has transformed the study of neural circuit function, but limitations in its application to species with large brains, such as non-human primates (NHPs), remain. A major challenge in NHP optogenetics is delivering light to sufficiently large volumes of deep neural tissue with high spatiotemporal precision, without simultaneously affecting superficial tissue. To overcome these limitations, we recently developed and tested in vivo in NHP cortex, the Utah Optrode Array (UOA). This is a 10×10 array of penetrating glass shanks, tiling a 4×4mm 2 area, bonded to interleaved needle-aligned and interstitial µLED arrays, which allows for independent photostimulation of deep and superficial brain tissue. Here, we investigate the acute biological response to UOA implantation in NHP cortex, with the goal of optimizing device design for reduced insertion trauma and subsequent chronic response. To this goal, we systematically vary UOA shank diameter, surface texture, tip geometry, and insertion pressure, and assess their effects on astrocytes, microglia, and neuronal viability, following acute implantation. We find that UOAs with shanks of smaller diameter, smooth surface texture and round tips cause the least damage. Higher insertion pressures have limited effects on the inflammatory response, but lead to greater tissue compression. Our results highlight the importance of balancing shank diameter, tip geometry, and insertion pressure in UOA design for preserving tissue integrity and improving long-term UOA performance and biocompatibility.
Collapse
|
8
|
Ghorpade KB, Agrawal S, Havelikar U. WITHDRAWN: Biomarker Detection and Therapy of Parkinson's and Alzheimer's disease using upconversion based approach: A Comprehensive Review. Ageing Res Rev 2025:102656. [PMID: 39788432 DOI: 10.1016/j.arr.2025.102656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
This article has been withdrawn at the request of the author(s). The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/policies/article-withdrawal.
Collapse
Affiliation(s)
- Kabirdas B Ghorpade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002 (Uttar Pradesh), India.
| | - Shivanshu Agrawal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002 (Uttar Pradesh), India
| | - Ujwal Havelikar
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India
| |
Collapse
|
9
|
Song Z, Sun Y, Liu P, Ruan H, He Y, Yin J, Xiao C, Ma J, Yu Y, Wang S, Gong Y, Lin ZW, Zhang Z, Chang C, Yang M. Terahertz Wave Alleviates Comorbidity Anxiety in Pain by Reducing the Binding Capacity of Nanostructured Glutamate Molecules to GluA2. RESEARCH (WASHINGTON, D.C.) 2024; 7:0535. [PMID: 39664293 PMCID: PMC11633831 DOI: 10.34133/research.0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024]
Abstract
Comorbid anxiety in chronic pain is clinically common, with a comorbidity rate of over 50%. The main treatments are based on pharmacological, interventional, and implantable approaches, which have limited efficacy and carry a risk of side effects. Here, we report a terahertz (THz, 1012 Hz) wave stimulation (THS) technique, which exerts nonthermal, long-term modulatory effects on neuronal activity by reducing the binding between nano-sized glutamate molecules and GluA2, leading to the relief of pain and comorbid anxiety-like behaviors in mice. In mice with co-occurring anxiety and chronic pain induced by complete Freund's adjuvant (CFA) injection, hyperactivity was observed in glutamatergic neurons in the anterior cingulate cortex (ACCGlu). Using whole-cell recording in ACC slices, we demonstrated that THS (34 THz) effectively inhibited the excitability of ACCGlu. Moreover, molecular dynamics simulations showed that THS reduced the number of hydrogen bonds bound between glutamate molecules and GluA2. Furthermore, THS target to the ACC in CFA-treatment mice suppressed ACCGlu hyperactivity and, as a result, alleviated pain and anxiety-like behaviors. Consistently, inhibition of ACCGlu hyperactivity by chemogenetics mimics THS-induced antinociceptive and antianxiety behavior. Together, our study provides evidence for THS as an intervention technique for modulating neuronal activity and a viable clinical treatment strategy for pain and comorbid anxiety.
Collapse
Affiliation(s)
- Zihua Song
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Yuankun Sun
- School of Electronic Science and Engineering,
University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Pan Liu
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Hao Ruan
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Yuanyuan He
- School of Safety Engineering,
North China Institute of Science and Technology, Hebei 065201, China
| | - Junkai Yin
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Chun Xiao
- School of Life Sciences,
Tsinghua University, Beijing 100081, China
| | - Jing Ma
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Yun Yu
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Life Science and Technology and Frontier Institute of Science and Technology,
Xi’an Jiaotong University, Xi’an 710049, China
| | - Shaomeng Wang
- School of Electronic Science and Engineering,
University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yubin Gong
- School of Electronic Science and Engineering,
University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Z. W. Lin
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Zhi Zhang
- Division of Life Sciences and Medicine, Department of Anesthesiology, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale,
University of Science and Technology of China, Hefei 230026, China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Physics,
Peking University, Beijing 100081, China
| | - Maojun Yang
- School of Life Sciences,
Tsinghua University, Beijing 100081, China
| |
Collapse
|
10
|
Andrews JP, Geng J, Voitiuk K, Elliott MAT, Shin D, Robbins A, Spaeth A, Wang A, Li L, Solis D, Keefe MG, Sevetson JL, Rivera de Jesús JA, Donohue KC, Larson HH, Ehrlich D, Auguste KI, Salama S, Sohal V, Sharf T, Haussler D, Cadwell CR, Schaffer DV, Chang EF, Teodorescu M, Nowakowski TJ. Multimodal evaluation of network activity and optogenetic interventions in human hippocampal slices. Nat Neurosci 2024; 27:2487-2499. [PMID: 39548326 DOI: 10.1038/s41593-024-01782-5] [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: 08/24/2023] [Accepted: 08/30/2024] [Indexed: 11/17/2024]
Abstract
Seizures are made up of the coordinated activity of networks of neurons, suggesting that control of neurons in the pathologic circuits of epilepsy could allow for control of the disease. Optogenetics has been effective at stopping seizure-like activity in non-human disease models by increasing inhibitory tone or decreasing excitation, although this effect has not been shown in human brain tissue. Many of the genetic means for achieving channelrhodopsin expression in non-human models are not possible in humans, and vector-mediated methods are susceptible to species-specific tropism that may affect translational potential. Here we demonstrate adeno-associated virus-mediated, optogenetic reductions in network firing rates of human hippocampal slices recorded on high-density microelectrode arrays under several hyperactivity-provoking conditions. This platform can serve to bridge the gap between human and animal studies by exploring genetic interventions on network activity in human brain tissue.
Collapse
Affiliation(s)
- John P Andrews
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jinghui Geng
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Kateryna Voitiuk
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Matthew A T Elliott
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - David Shin
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Ash Robbins
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Alex Spaeth
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Albert Wang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Lin Li
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Daniel Solis
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Matthew G Keefe
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica L Sevetson
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, USA
| | | | - Kevin C Donohue
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - H Hanh Larson
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Drew Ehrlich
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Computational Media, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Kurtis I Auguste
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sofie Salama
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, USA
| | - Vikaas Sohal
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tal Sharf
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - David Haussler
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Cathryn R Cadwell
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - David V Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, California, CA, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute (IGI), University of California, Berkeley, Berkeley, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Mircea Teodorescu
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA.
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA.
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, USA.
| | - Tomasz Jan Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Uji M, Kondo J, Hara-Miyauchi C, Akimoto S, Haruki R, Sasaki Y, Kimizuka N, Ajioka I, Yanai N. In Vivo Optogenetics Based on Heavy Metal-Free Photon Upconversion Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405509. [PMID: 39308228 DOI: 10.1002/adma.202405509] [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: 04/17/2024] [Revised: 09/14/2024] [Indexed: 11/16/2024]
Abstract
Photon upconversion (UC) from red or near-infrared (NIR) light to blue light is promising for in vivo optogenetics. However, the examples of in vivo optogenetics have been limited to lanthanide inorganic UC nanoparticles, and there have been no examples of optogenetics without using heavy metals. Here the first example of in vivo optogenetics using biocompatible heavy metal-free TTA-UC nanoemulsions is shown. A new organic TADF sensitizer, a boron difluoride curcuminoid derivative modified with a bromo group, can promote intersystem crossing to the excited triplet state, significantly improving TTA-UC efficiency. The TTA-UC nanoparticles formed from biocompatible surfactants and methyl oleate acquire water dispersibility and remarkable oxygen tolerance. By combining with genome engineering technology using the blue light-responding photoactivatable Cre-recombinase (PA-Cre), TTA-UC nanoparticles promote Cre-reporter EGFP expression in neurons in vitro and in vivo. The results open new opportunities toward deep-tissue control of neural activities based on heavy metal-free fully organic UC systems.
Collapse
Affiliation(s)
- Masanori Uji
- Department of Applied Chemistry, Graduate School of Engineering and Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Jumpei Kondo
- Department of Applied Chemistry, Graduate School of Engineering and Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Chikako Hara-Miyauchi
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), 3-2-1 Sakato, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-0012, Japan
| | - Saori Akimoto
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), 3-2-1 Sakato, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-0012, Japan
| | - Rena Haruki
- Department of Applied Chemistry, Graduate School of Engineering and Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yoichi Sasaki
- Department of Applied Chemistry, Graduate School of Engineering and Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Nobuo Kimizuka
- Department of Applied Chemistry, Graduate School of Engineering and Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Itsuki Ajioka
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Kanagawa Institute of Industrial Science and Technology (KISTEC), 3-2-1 Sakato, Takatsu-ku, Kawasaki-shi, Kanagawa, 213-0012, Japan
- Research Center for Autonomous Systems Materialogy (AsMAT), Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama-shi, Kanagawa, 226-8501, Japan
| | - Nobuhiro Yanai
- Department of Applied Chemistry, Graduate School of Engineering and Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
- CREST, JST, 4-1-8 Honcho, Kawaguchi-shi, Saitama, 332-0012, Japan
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
12
|
Lanni I, Chiacchierini G, Papagno C, Santangelo V, Campolongo P. Treating Alzheimer's disease with brain stimulation: From preclinical models to non-invasive stimulation in humans. Neurosci Biobehav Rev 2024; 165:105831. [PMID: 39074672 DOI: 10.1016/j.neubiorev.2024.105831] [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: 05/20/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) is a severe and progressive neurodegenerative condition that exerts detrimental effects on brain function. As of now, there is no effective treatment for AD patients. This review explores two distinct avenues of research. The first revolves around the use of animal studies and preclinical models to gain insights into AD's underlying mechanisms and potential treatment strategies. Specifically, it delves into the effectiveness of interventions such as Optogenetics and Chemogenetics, shedding light on their implications for understanding pathophysiological mechanisms and potential therapeutic applications. The second avenue focuses on non-invasive brain stimulation (NiBS) techniques in the context of AD. Evidence suggests that NiBS can successfully modulate cognitive functions associated with various neurological and neuropsychiatric disorders, including AD, as demonstrated by promising findings. Here, we critically assessed recent findings in AD research belonging to these lines of research and discuss their potential impact on the clinical horizon of AD treatment. These multifaceted approaches offer hope for advancing our comprehension of AD pathology and developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Ilenia Lanni
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Chiacchierini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Costanza Papagno
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Valerio Santangelo
- Functional Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
13
|
Bolonduro OA, Chen Z, Fucetola CP, Lai YR, Cote M, Kajola RO, Rao AA, Liu H, Tzanakakis ES, Timko BP. An Integrated Optogenetic and Bioelectronic Platform for Regulating Cardiomyocyte Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402236. [PMID: 39054679 PMCID: PMC11423186 DOI: 10.1002/advs.202402236] [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: 03/01/2024] [Revised: 06/24/2024] [Indexed: 07/27/2024]
Abstract
Bioelectronic medicine is emerging as a powerful approach for restoring lost endogenous functions and addressing life-altering maladies such as cardiac disorders. Systems that incorporate both modulation of cellular function and recording capabilities can enhance the utility of these approaches and their customization to the needs of each patient. Here we report an integrated optogenetic and bioelectronic platform for stable and long-term stimulation and monitoring of cardiomyocyte function in vitro. Optical inputs are achieved through the expression of a photoactivatable adenylyl cyclase, that when irradiated with blue light causes a dose-dependent and time-limited increase in the secondary messenger cyclic adenosine monophosphate with subsequent rise in autonomous cardiomyocyte beating rate. Bioelectronic readouts are obtained through a multi-electrode array that measures real-time electrophysiological responses at 32 spatially-distinct locations. Irradiation at 27 µW mm-2 results in a 14% elevation of the beating rate within 20-25 min, which remains stable for at least 2 h. The beating rate can be cycled through "on" and "off" light states, and its magnitude is a monotonic function of irradiation intensity. The integrated platform can be extended to stretchable and flexible substrates, and can open new avenues in bioelectronic medicine, including closed-loop systems for cardiac regulation and intervention, for example, in the context of arrythmias.
Collapse
Affiliation(s)
| | - Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
| | - Corey P Fucetola
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Yan-Ru Lai
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Megan Cote
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Rofiat O Kajola
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Akshita A Rao
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Haitao Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China
| | - Emmanuel S Tzanakakis
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
- Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Brian P Timko
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
14
|
Li L, Zhang B, Zhao W, Sheng D, Yin L, Sheng X, Yao D. Multimodal Technologies for Closed-Loop Neural Modulation and Sensing. Adv Healthc Mater 2024; 13:e2303289. [PMID: 38640468 DOI: 10.1002/adhm.202303289] [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: 09/27/2023] [Revised: 03/11/2024] [Indexed: 04/21/2024]
Abstract
Existing methods for studying neural circuits and treating neurological disorders are typically based on physical and chemical cues to manipulate and record neural activities. These approaches often involve predefined, rigid, and unchangeable signal patterns, which cannot be adjusted in real time according to the patient's condition or neural activities. With the continuous development of neural interfaces, conducting in vivo research on adaptive and modifiable treatments for neurological diseases and neural circuits is now possible. In this review, current and potential integration of various modalities to achieve precise, closed-loop modulation, and sensing in neural systems are summarized. Advanced materials, devices, or systems that generate or detect electrical, magnetic, optical, acoustic, or chemical signals are highlighted and utilized to interact with neural cells, tissues, and networks for closed-loop interrogation. Further, the significance of developing closed-loop techniques for diagnostics and treatment of neurological disorders such as epilepsy, depression, rehabilitation of spinal cord injury patients, and exploration of brain neural circuit functionality is elaborated.
Collapse
Affiliation(s)
- Lizhu Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Bozhen Zhang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Wenxin Zhao
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - David Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Dezhong Yao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
15
|
Li Y, Lee SH, Yu C, Hsu LM, Wang TWW, Do K, Kim HJ, Shih YYI, Grill WM. Optogenetic fMRI reveals therapeutic circuits of subthalamic nucleus deep brain stimulation. Brain Stimul 2024; 17:947-957. [PMID: 39096961 PMCID: PMC11364984 DOI: 10.1016/j.brs.2024.07.022] [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: 04/12/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
While deep brain stimulation (DBS) is widely employed for managing motor symptoms in Parkinson's disease (PD), its exact circuit mechanisms remain controversial. To identify the neural targets affected by therapeutic DBS in PD, we analyzed DBS-evoked whole brain activity in female hemi-parkinsonian rats using functional magnetic resonance imaging (fMRI). We delivered subthalamic nucleus (STN) DBS at various stimulation pulse repetition rates using optogenetics, allowing unbiased examination of cell-type specific STN feedforward neural activity. Unilateral optogenetic STN DBS elicited pulse repetition rate-dependent alterations of blood-oxygenation-level-dependent (BOLD) signals in SNr (substantia nigra pars reticulata), GP (globus pallidus), and CPu (caudate putamen). Notably, this modulation effectively ameliorated pathological circling behavior in animals expressing the kinetically faster Chronos opsin, but not in animals expressing ChR2. Furthermore, mediation analysis revealed that the pulse repetition rate-dependent behavioral rescue was significantly mediated by optogenetic DBS induced activity changes in GP and CPu, but not in SNr. This suggests that the activation of GP and CPu are critically involved in the therapeutic mechanisms of STN DBS.
Collapse
Affiliation(s)
- Yuhui Li
- Department of Biomedical Engineering, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Chunxiu Yu
- Department of Biomedical Engineering, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Tzu-Wen W Wang
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Khoa Do
- Department of Biomedical Engineering, USA
| | - Hyeon-Joong Kim
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA; Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC, USA.
| | - Warren M Grill
- Department of Biomedical Engineering, USA; Department of Electrical and Computer Engineering, USA; Department of Neurobiology, Duke University, Durham, NC, USA; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
16
|
De Ridder D, Siddiqi MA, Dauwels J, Serdijn WA, Strydis C. NeuroDots: From Single-Target to Brain-Network Modulation: Why and What Is Needed? Neuromodulation 2024; 27:711-729. [PMID: 38639704 DOI: 10.1016/j.neurom.2024.01.003] [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: 08/07/2023] [Revised: 11/05/2023] [Accepted: 01/10/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVES Current techniques in brain stimulation are still largely based on a phrenologic approach that a single brain target can treat a brain disorder. Nevertheless, meta-analyses of brain implants indicate an overall success rate of 50% improvement in 50% of patients, irrespective of the brain-related disorder. Thus, there is still a large margin for improvement. The goal of this manuscript is to 1) develop a general theoretical framework of brain functioning that is amenable to surgical neuromodulation, and 2) describe the engineering requirements of the next generation of implantable brain stimulators that follow from this theoretic model. MATERIALS AND METHODS A neuroscience and engineering literature review was performed to develop a universal theoretical model of brain functioning and dysfunctioning amenable to surgical neuromodulation. RESULTS Even though a single target can modulate an entire network, research in network science reveals that many brain disorders are the consequence of maladaptive interactions among multiple networks rather than a single network. Consequently, targeting the main connector hubs of those multiple interacting networks involved in a brain disorder is theoretically more beneficial. We, thus, envision next-generation network implants that will rely on distributed, multisite neuromodulation targeting correlated and anticorrelated interacting brain networks, juxtaposing alternative implant configurations, and finally providing solid recommendations for the realization of such implants. In doing so, this study pinpoints the potential shortcomings of other similar efforts in the field, which somehow fall short of the requirements. CONCLUSION The concept of network stimulation holds great promise as a universal approach for treating neurologic and psychiatric disorders.
Collapse
Affiliation(s)
- Dirk De Ridder
- Section of Neurosurgery, Department of Surgical Sciences, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.
| | - Muhammad Ali Siddiqi
- Department of Electrical Engineering, Lahore University of Management Sciences, Lahore, Pakistan; Neuroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands; Quantum and Computer Engineering Department, Delft University of Technology, Delft, The Netherlands
| | - Justin Dauwels
- Microelectronics Department, Delft University of Technology, Delft, The Netherlands
| | - Wouter A Serdijn
- Neuroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands; Section Bioelectronics, Delft University of Technology, Delft, The Netherlands
| | - Christos Strydis
- Neuroscience Department, Erasmus Medical Center, Rotterdam, The Netherlands; Quantum and Computer Engineering Department, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
17
|
El Hajj R, Al Sagheer T, Ballout N. Optogenetics in chronic neurodegenerative diseases, controlling the brain with light: A systematic review. J Neurosci Res 2024; 102:e25321. [PMID: 38588013 DOI: 10.1002/jnr.25321] [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: 10/24/2023] [Revised: 02/20/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024]
Abstract
Neurodegenerative diseases are progressive disorders characterized by synaptic loss and neuronal death. Optogenetics combines optical and genetic methods to control the activity of specific cell types. The efficacy of this approach in neurodegenerative diseases has been investigated in many reviews, however, none of them tackled it systematically. Our study aimed to review systematically the findings of optogenetics and its potential applications in animal models of chronic neurodegenerative diseases and compare it with deep brain stimulation and designer receptors exclusively activated by designer drugs techniques. The search strategy was performed based on the PRISMA guidelines and the risk of bias was assessed following the Systematic Review Centre for Laboratory Animal Experimentation tool. A total of 247 articles were found, of which 53 were suitable for the qualitative analysis. Our data revealed that optogenetic manipulation of distinct neurons in the brain is efficient in rescuing memory impairment, alleviating neuroinflammation, and reducing plaque pathology in Alzheimer's disease. Similarly, this technique shows an advanced understanding of the contribution of various neurons involved in the basal ganglia pathways with Parkinson's disease motor symptoms and pathology. However, the optogenetic application using animal models of Huntington's disease, multiple sclerosis, and amyotrophic lateral sclerosis was limited. Optogenetics is a promising technique that enhanced our knowledge in the research of neurodegenerative diseases and addressed potential therapeutic solutions for managing these diseases' symptoms and delaying their progression. Nevertheless, advanced investigations should be considered to improve optogenetic tools' efficacy and safety to pave the way for their translatability to the clinic.
Collapse
Affiliation(s)
- Rojine El Hajj
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Tareq Al Sagheer
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Nissrine Ballout
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| |
Collapse
|
18
|
Sakurai K. Rethinking c-Fos for understanding drug action in the brain. J Biochem 2024; 175:377-381. [PMID: 38153290 DOI: 10.1093/jb/mvad110] [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: 09/04/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 12/29/2023] Open
Abstract
Understanding the mechanisms of drug action in the brain, from the genetic to the neural circuit level, is crucial for the development of new agents that act upon the central nervous system. Determining the brain regions and neurons affected by a drug is essential for revealing its mechanism of action in the brain. c-Fos, a marker of neuronal activation, has been widely used to detect neurons activated by stimuli with high spatial resolution. In this review, the use of c-Fos for the visualization and manipulation of activated neurons is introduced. I also explain that a higher temporal resolution can be achieved by changing the staining method for visualization of c-Fos. Moreover, a new method that allows labeling and manipulating commonly activated neurons using two different stimuli is proposed.
Collapse
Affiliation(s)
- Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
19
|
Cho M, Han JK, Suh J, Kim JJ, Ryu JR, Min IS, Sang M, Lim S, Kim TS, Kim K, Kang K, Hwang K, Kim K, Hong EB, Nam MH, Kim J, Song YM, Lee GJ, Cho IJ, Yu KJ. Fully bioresorbable hybrid opto-electronic neural implant system for simultaneous electrophysiological recording and optogenetic stimulation. Nat Commun 2024; 15:2000. [PMID: 38448437 PMCID: PMC10917781 DOI: 10.1038/s41467-024-45803-0] [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: 04/20/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Bioresorbable neural implants based on emerging classes of biodegradable materials offer a promising solution to the challenges of secondary surgeries for removal of implanted devices required for existing neural implants. In this study, we introduce a fully bioresorbable flexible hybrid opto-electronic system for simultaneous electrophysiological recording and optogenetic stimulation. The flexible and soft device, composed of biodegradable materials, has a direct optical and electrical interface with the curved cerebral cortex surface while exhibiting excellent biocompatibility. Optimized to minimize light transmission losses and photoelectric artifact interference, the device was chronically implanted in the brain of transgenic mice and performed to photo-stimulate the somatosensory area while recording local field potentials. Thus, the presented hybrid neural implant system, comprising biodegradable materials, promises to provide monitoring and therapy modalities for versatile applications in biomedicine.
Collapse
Affiliation(s)
- Myeongki Cho
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong-Kyu Han
- Brain Science Institute, Korea Institute of Science and Technology, 5. Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jungmin Suh
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong Jin Kim
- Department of Electronics Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| | - Jae Ryun Ryu
- Department of Anatomy, College of Medicine, Korea University, 17-gil Koryodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - In Sik Min
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Mingyu Sang
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Selin Lim
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Tae Soo Kim
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyubeen Kim
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyowon Kang
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyuhyun Hwang
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kanghwan Kim
- Brain Science Institute, Korea Institute of Science and Technology, 5. Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Eun-Bin Hong
- Center for Brain Function, Korea Institute of Science and Technology 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Min-Ho Nam
- Center for Brain Function, Korea Institute of Science and Technology 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jongbaeg Kim
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Min Song
- School of Electrical Engineering and Computer Science (EECS), Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Gil Ju Lee
- Department of Electronics Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.
| | - Il-Joo Cho
- Department of Convergence Medicine, College of Medicine, Korea University, 17-gil Koryodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
- Department of Anatomy, College of Medicine, Korea University, 7-gil Koryodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Ki Jun Yu
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Department of Electrical and Electronic Engineering, YU-Korea Institute of Science and Technology (KIST) Institute, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
20
|
Li Y, Lee SH, Yu C, Hsu LM, Wang TWW, Do K, Kim HJ, Shih YYI, Grill WM. Optogenetic fMRI reveals therapeutic circuits of subthalamic nucleus deep brain stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581627. [PMID: 38464010 PMCID: PMC10925223 DOI: 10.1101/2024.02.22.581627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
While deep brain stimulation (DBS) is widely employed for managing motor symptoms in Parkinson's disease (PD), its exact circuit mechanisms remain controversial. To identify the neural targets affected by therapeutic DBS in PD, we analyzed DBS-evoked whole brain activity in female hemi-parkinsonian rats using function magnetic resonance imaging (fMRI). We delivered subthalamic nucleus (STN) DBS at various stimulation pulse repetition rates using optogenetics, allowing unbiased examinations of cell-type specific STN feed-forward neural activity. Unilateral STN optogenetic stimulation elicited pulse repetition rate-dependent alterations of blood-oxygenation-level-dependent (BOLD) signals in SNr (substantia nigra pars reticulata), GP (globus pallidus), and CPu (caudate putamen). Notably, these manipulations effectively ameliorated pathological circling behavior in animals expressing the kinetically faster Chronos opsin, but not in animals expressing ChR2. Furthermore, mediation analysis revealed that the pulse repetition rate-dependent behavioral rescue was significantly mediated by optogenetically induced activity changes in GP and CPu, but not in SNr. This suggests that the activation of GP and CPu are critically involved in the therapeutic mechanisms of STN DBS.
Collapse
|
21
|
Maloney R, Quattrochi L, Yoon J, Souza R, Berson D. Efficacy and specificity of melanopsin reporters for retinal ganglion cells. J Comp Neurol 2024; 532:e25591. [PMID: 38375612 PMCID: PMC11000424 DOI: 10.1002/cne.25591] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/03/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are specialized retinal output neurons that mediate behavioral, neuroendocrine, and developmental responses to environmental light. There are diverse molecular strategies for marking ipRGCs, especially in mice, making them among the best characterized retinal ganglion cells (RGCs). With the development of more sensitive reporters, new subtypes of ipRGCs have emerged. We therefore tested high-sensitivity reporter systems to see whether we could reveal yet more. Substantial confusion remains about which of the available methods, if any, label all and only ipRGCs. Here, we compared many different methods for labeling of ipRGCs, including anti-melanopsin immunofluorescence, Opn4-GFP BAC transgenic mice, and Opn4cre mice crossed with three different Cre-specific reporters (Z/EG, Ai9, and Ai14) or injected with Cre-dependent (DIO) AAV2. We show that Opn4cre mice, when crossed with sensitive Cre-reporter mice, label numerous ganglion cell types that lack intrinsic photosensitivity. Though other methods label ipRGCs specifically, they do not label the entire population of ipRGCs. We conclude that no existing method labels all and only ipRGCs. We assess the appropriateness of each reporter for particular applications and integrate findings across reporters to estimate that the overall abundance of ipRGCs among mouse RGCs may approach 11%.
Collapse
Affiliation(s)
- Ryan Maloney
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Lauren Quattrochi
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - James Yoon
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - Rachel Souza
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - David Berson
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
22
|
Withey SL, Pizzagalli DA, Bergman J. Translational In Vivo Assays in Behavioral Biology. Annu Rev Pharmacol Toxicol 2024; 64:435-453. [PMID: 37708432 DOI: 10.1146/annurev-pharmtox-051921-093711] [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: 09/16/2023]
Abstract
The failure of preclinical research to advance successful candidate medications in psychiatry has created a paradigmatic crisis in psychiatry. The Research Domain Criteria (RDoC) initiative was designed to remedy this situation with a neuroscience-based approach that employs multimodal and cross-species in vivo methodology to increase the probability of translational findings and, consequently, drug discovery. The present review underscores the feasibility of this methodological approach by briefly reviewing, first, the use of multidimensional and cross-species methodologies in traditional behavioral pharmacology and, subsequently, the utility of this approach in contemporary neuroimaging and electrophysiology research-with a focus on the value of functionally homologous studies in nonhuman and human subjects. The final section provides a brief review of the RDoC, with a focus on the potential strengths and weaknesses of its domain-based underpinnings. Optimistically, this mechanistic and multidimensional approach in neuropsychiatric research will lead to novel therapeutics for the management of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sarah L Withey
- Preclinical Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts, USA;
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- McLean Imaging Center, McLean Hospital, Belmont, Massachusetts, USA
| | - Diego A Pizzagalli
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- McLean Imaging Center, McLean Hospital, Belmont, Massachusetts, USA
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts, USA
| | - Jack Bergman
- Preclinical Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts, USA;
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Li M, Yang G. A mesocortical glutamatergic pathway modulates neuropathic pain independent of dopamine co-release. Nat Commun 2024; 15:643. [PMID: 38245542 PMCID: PMC10799877 DOI: 10.1038/s41467-024-45035-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
Dysfunction in the mesocortical pathway, connecting the ventral tegmental area (VTA) to the prefrontal cortex, has been implicated in chronic pain. While extensive research has focused on the role of dopamine, the contribution of glutamatergic signaling in pain modulation remains unknown. Using in vivo calcium imaging, we observe diminished VTA glutamatergic activity targeting the prelimbic cortex (PL) in a mouse model of neuropathic pain. Optogenetic activation of VTA glutamatergic terminals in the PL alleviates neuropathic pain, whereas inhibiting these terminals in naïve mice induces pain-like responses. Importantly, this pain-modulating effect is independent of dopamine co-release, as demonstrated by CRISPR/Cas9-mediated gene deletion. Furthermore, we show that VTA neurons primarily project to excitatory neurons in the PL, and their activation restores PL outputs to the anterior cingulate cortex, a key region involved in pain processing. These findings reveal a distinct mesocortical glutamatergic pathway that critically modulates neuropathic pain independent of dopamine signaling.
Collapse
Affiliation(s)
- Miao Li
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
24
|
Jiang S, Wu X, Yang F, Rommelfanger NJ, Hong G. Activation of mechanoluminescent nanotransducers by focused ultrasound enables light delivery to deep-seated tissue in vivo. Nat Protoc 2023; 18:3787-3820. [PMID: 37914782 PMCID: PMC11405139 DOI: 10.1038/s41596-023-00895-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/27/2023] [Indexed: 11/03/2023]
Abstract
Light is used extensively in biological and medical research for optogenetic neuromodulation, fluorescence imaging, photoactivatable gene editing and light-based therapies. The major challenge to the in vivo implementation of light-based methods in deep-seated structures of the brain or of internal organs is the limited penetration of photons in biological tissue. The presence of light scattering and absorption has resulted in the development of invasive techniques such as the implantation of optical fibers, the insertion of endoscopes and the surgical removal of overlying tissues to overcome light attenuation and deliver it deep into the body. However, these procedures are highly invasive and make it difficult to reposition and adjust the illuminated area in each animal. Here, we detail a noninvasive approach to deliver light (termed 'deLight') in deep tissue via systemically injected mechanoluminescent nanotransducers that can be gated by using focused ultrasound. This approach achieves localized light emission with sub-millimeter resolution and millisecond response times in any vascularized organ of living mice without requiring invasive implantation of light-emitting devices. For example, deLight enables optogenetic neuromodulation in live mice without a craniotomy or brain implants. deLight provides a generalized method for applications that require a light source in deep tissues in vivo, such as deep-brain fluorescence imaging and photoactivatable genome editing. The implementation of the entire protocol for an in vivo application takes ~1-2 weeks.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Xiang Wu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Fan Yang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Nicholas J Rommelfanger
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Stierschneider A, Wiesner C. Shedding light on the molecular and regulatory mechanisms of TLR4 signaling in endothelial cells under physiological and inflamed conditions. Front Immunol 2023; 14:1264889. [PMID: 38077393 PMCID: PMC10704247 DOI: 10.3389/fimmu.2023.1264889] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Toll-like receptor 4 (TLR4) are part of the innate immune system. They are capable of recognizing pathogen-associated molecular patterns (PAMPS) of microbes, and damage-associated molecular patterns (DAMPs) of damaged tissues. Activation of TLR4 initiates downstream signaling pathways that trigger the secretion of cytokines, type I interferons, and other pro-inflammatory mediators that are necessary for an immediate immune response. However, the systemic release of pro-inflammatory proteins is a powerful driver of acute and chronic inflammatory responses. Over the past decades, immense progress has been made in clarifying the molecular and regulatory mechanisms of TLR4 signaling in inflammation. However, the most common strategies used to study TLR4 signaling rely on genetic manipulation of the TLR4 or the treatment with agonists such as lipopolysaccharide (LPS) derived from the outer membrane of Gram-negative bacteria, which are often associated with the generation of irreversible phenotypes in the target cells or unintended cytotoxicity and signaling crosstalk due to off-target or pleiotropic effects. Here, optogenetics offers an alternative strategy to control and monitor cellular signaling in an unprecedented spatiotemporally precise, dose-dependent, and non-invasive manner. This review provides an overview of the structure, function and signaling pathways of the TLR4 and its fundamental role in endothelial cells under physiological and inflammatory conditions, as well as the advances in TLR4 modulation strategies.
Collapse
Affiliation(s)
| | - Christoph Wiesner
- Department Science & Technology, Institute Biotechnology, IMC Krems University of Applied Sciences, Krems, Austria
| |
Collapse
|
26
|
Seo H, Chung WG, Kwon YW, Kim S, Hong YM, Park W, Kim E, Lee J, Lee S, Kim M, Lim K, Jeong I, Song H, Park JU. Smart Contact Lenses as Wearable Ophthalmic Devices for Disease Monitoring and Health Management. Chem Rev 2023; 123:11488-11558. [PMID: 37748126 PMCID: PMC10571045 DOI: 10.1021/acs.chemrev.3c00290] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Indexed: 09/27/2023]
Abstract
The eye contains a complex network of physiological information and biomarkers for monitoring disease and managing health, and ocular devices can be used to effectively perform point-of-care diagnosis and disease management. This comprehensive review describes the target biomarkers and various diseases, including ophthalmic diseases, metabolic diseases, and neurological diseases, based on the physiological and anatomical background of the eye. This review also includes the recent technologies utilized in eye-wearable medical devices and the latest trends in wearable ophthalmic devices, specifically smart contact lenses for the purpose of disease management. After introducing other ocular devices such as the retinal prosthesis, we further discuss the current challenges and potential possibilities of smart contact lenses.
Collapse
Affiliation(s)
- Hunkyu Seo
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Won Gi Chung
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Yong Won Kwon
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Sumin Kim
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Yeon-Mi Hong
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Wonjung Park
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Enji Kim
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Jakyoung Lee
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Sanghoon Lee
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Moohyun Kim
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Kyeonghee Lim
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Inhea Jeong
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Hayoung Song
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
| | - Jang-Ung Park
- Department
of Materials Science and Engineering, Yonsei
University, Seoul 03722, Republic
of Korea
- Department
of Neurosurgery, Yonsei University College
of Medicine, Seoul 03722, Republic of Korea
- Center
for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul 03722, Republic
of Korea
| |
Collapse
|
27
|
Xu S, Momin M, Ahmed S, Hossain A, Veeramuthu L, Pandiyan A, Kuo CC, Zhou T. Illuminating the Brain: Advances and Perspectives in Optoelectronics for Neural Activity Monitoring and Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303267. [PMID: 37726261 DOI: 10.1002/adma.202303267] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/30/2023] [Indexed: 09/21/2023]
Abstract
Optogenetic modulation of brain neural activity that combines optical and electrical modes in a unitary neural system has recently gained robust momentum. Controlling illumination spatial coverage, designing light-activated modulators, and developing wireless light delivery and data transmission are crucial for maximizing the use of optical neuromodulation. To this end, biocompatible electrodes with enhanced optoelectrical performance, device integration for multiplexed addressing, wireless transmission, and multimodal operation in soft systems have been developed. This review provides an outlook for uniformly illuminating large brain areas while spatiotemporally imaging the neural responses upon optoelectrical stimulation with little artifacts. Representative concepts and important breakthroughs, such as head-mounted illumination, multiple implanted optical fibers, and micro-light-delivery devices, are discussed. Examples of techniques that incorporate electrophysiological monitoring and optoelectrical stimulation are presented. Challenges and perspectives are posed for further research efforts toward high-density optoelectrical neural interface modulation, with the potential for nonpharmacological neurological disease treatments and wireless optoelectrical stimulation.
Collapse
Affiliation(s)
- Shumao Xu
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Marzia Momin
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Salahuddin Ahmed
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Arafat Hossain
- Department of Electrical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Loganathan Veeramuthu
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Archana Pandiyan
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Chi-Ching Kuo
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Tao Zhou
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| |
Collapse
|
28
|
Di Vito A, Amiri P, Bornemann S, Schöttler G, Vergin M, Meierhofer F, Gülink J, Waag A, Canals J, Diéguez A, Prades JD, Auf der Maur M. Design study of a micro illumination platform based on GaN microLED arrays. APPLIED OPTICS 2023; 62:7503-7511. [PMID: 37855520 DOI: 10.1364/ao.498432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/06/2023] [Indexed: 10/20/2023]
Abstract
The design study of a micro illumination tool based on GaN microLED arrays is presented. The high spatio-temporal resolution and the capability of generating fully customized optical patterns that characterize the proposed platform would enable the manipulation of biological systems, e.g., for optogenetics applications. Based on ray tracing simulations, the design aspects that mainly affect the device performance have been identified, and the related structural parameters have been optimized to improve the extraction efficiency and the spatial resolution of the resulting light patterns. Assuming that the device is a bottom emitter, and the light is extracted from the n-side, the presence of mesa-structures on the p-side of the GaN layer can affect both the efficiency and the resolution, being optimized for different values of the mesa-side inclination angle. The full width at half maximum (FWHM) of the extracted spots is mainly determined by the substrate thickness, and the relation between the FWHM and the array pitch represents a criterion to define the resolution. Namely, when F W H M
Collapse
|
29
|
Kimura K, Nagai Y, Hatanaka G, Fang Y, Tanabe S, Zheng A, Fujiwara M, Nakano M, Hori Y, Takeuchi RF, Inagaki M, Minamimoto T, Fujita I, Inoue KI, Takada M. A mosaic adeno-associated virus vector as a versatile tool that exhibits high levels of transgene expression and neuron specificity in primate brain. Nat Commun 2023; 14:4762. [PMID: 37553329 PMCID: PMC10409865 DOI: 10.1038/s41467-023-40436-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
Recent emphasis has been placed on gene transduction mediated through recombinant adeno-associated virus (AAV) vector to manipulate activity of neurons and their circuitry in the primate brain. In the present study, we created a novel vector of which capsid was composed of capsid proteins derived from both of the AAV serotypes 1 and 2 (AAV1 and AAV2). Following the injection into the frontal cortex of macaque monkeys, this mosaic vector, termed AAV2.1 vector, was found to exhibit the excellence in transgene expression (for AAV1 vector) and neuron specificity (for AAV2 vector) simultaneously. To explore its applicability to chemogenetic manipulation and in vivo calcium imaging, the AAV2.1 vector expressing excitatory DREADDs or GCaMP was injected into the striatum or the visual cortex of macaque monkeys, respectively. Our results have defined that such vectors secure intense and stable expression of the target proteins and yield conspicuous modulation and imaging of neuronal activity.
Collapse
Affiliation(s)
- Kei Kimura
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Yuji Nagai
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Gaku Hatanaka
- Laboratory for Cognitive Neuroscience, Graduate School of Frontier Biosciences, Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yang Fang
- Laboratory for Cognitive Neuroscience, Graduate School of Frontier Biosciences, Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Soshi Tanabe
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Andi Zheng
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Maki Fujiwara
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Mayuko Nakano
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Yukiko Hori
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Ryosuke F Takeuchi
- Laboratory for Cognitive Neuroscience, Graduate School of Frontier Biosciences, Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mikio Inagaki
- Laboratory for Cognitive Neuroscience, Graduate School of Frontier Biosciences, Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takafumi Minamimoto
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Ichiro Fujita
- Laboratory for Cognitive Neuroscience, Graduate School of Frontier Biosciences, Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology and Osaka University, 1-4 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan.
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama, 332-0012, Japan.
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan.
| |
Collapse
|
30
|
Margalit SN, Slovin H. Spatio-temporal activation patterns of neuronal population evoked by optostimulation and the comparison to electrical microstimulation. Sci Rep 2023; 13:12689. [PMID: 37542091 PMCID: PMC10403613 DOI: 10.1038/s41598-023-39808-w] [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: 12/26/2022] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
Optostimulation and electrical microstimulation are well-established techniques that enable to artificially stimulate the brain. While the activation patterns evoked by microstimulation in cortical network are well characterized, much less is known for optostimulation. Specifically, the activation maps of neuronal population at the membrane potential level and direct measurements of these maps were barely reported. In addition, only a few studies compared the activation patterns evoked by microstimulation and optostimulation. In this study we addressed these issues by applying optostimulation in the barrel cortex of anesthetized rats after a short (ShortExp) or a long (LongExp) opsin expression time and compared it to microstimulation. We measured the membrane potential of neuronal populations at high spatial (meso-scale) and temporal resolution using voltage-sensitive dye imaging. Longer optostimulation pulses evoked higher neural responses spreading over larger region relative to short pulses. Interestingly, similar optostimulation pulses evoked stronger and more prolonged population response in the LongExp vs. the ShortExp condition. Finally, the spatial activation patterns evoked in the LongExp condition showed an intermediate state, with higher resemblance to the microstimulation at the stimulation site. Therefore, short microstimulation and optostimulation can induce wide spread activation, however the effects of optostimulation depend on the opsin expression time.
Collapse
Affiliation(s)
| | - Hamutal Slovin
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel.
| |
Collapse
|
31
|
Zhao J, Yu Y, Han F, Wang Q. Regulating epileptiform discharges by heterogeneous interneurons in thalamocortical model. CHAOS (WOODBURY, N.Y.) 2023; 33:083128. [PMID: 37561121 DOI: 10.1063/5.0163243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023]
Abstract
Inhibitory interneurons in the cortex are abundant and have diverse roles, classified as parvalbumin (PV), somatostatin (SOM), and vasoactive intestinal polypeptide (VIP) according to chemically defined categories. Currently, their involvement with seizures has been partially uncovered in physiological terms. Here, we propose a corticothalamic model containing heterogeneous interneurons to study the effects of various interneurons on absence seizure dynamics by means of optogenetic stimulation. First, the important role of feedforward inhibition caused by SRN→PV→PN projections on seizures is verified. Then, we demonstrate that light activation targeting either PV or SOM INs can control seizures. Finally, with different inhibition contributions from PV INs and SOM INs, the possible disinhibitory effect of blue light acting on VIP INs is mainly discussed. The results suggest that depending on the inhibition degree of both types, the disinhibition brought about by the VIP INs will trigger seizures, will control seizures, and will not work or cause the PNs to tend toward a high saturation state with high excitability. The circuit mechanism and the related bifurcation characteristics in various cases are emphatically revealed. In the model presented, in addition to Hopf and saddle-node bifurcations, the system may also undergo period-doubling and torus bifurcations under stimulus action, with more complex dynamics. Our work may provide a theoretical basis for understanding and further exploring the role of heterogeneous interneurons, in particular, the VIP INs, a novel target, in absence seizures.
Collapse
Affiliation(s)
- Jinyi Zhao
- Department of Dynamics and Control, Beihang University, Beijing 100191, China
| | - Ying Yu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Fang Han
- College of Information Science and Technology, Donghua University, Shanghai 201620, China
| | - Qingyun Wang
- Department of Dynamics and Control, Beihang University, Beijing 100191, China
| |
Collapse
|
32
|
Ding L, Balsamo G, Diamantaki M, Preston-Ferrer P, Burgalossi A. Opto-juxtacellular interrogation of neural circuits in freely moving mice. Nat Protoc 2023; 18:2415-2440. [PMID: 37420087 DOI: 10.1038/s41596-023-00842-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/11/2023] [Indexed: 07/09/2023]
Abstract
Neural circuits are assembled from an enormous variety of neuronal cell types. Although significant advances have been made in classifying neurons on the basis of morphological, molecular and electrophysiological properties, understanding how this diversity contributes to brain function during behavior has remained a major experimental challenge. Here, we present an extension to our previous protocol, in which we describe the technical procedures for performing juxtacellular opto-tagging of single neurons in freely moving mice by using Channelrhodopsin-2-expressing viral vectors. This method allows one to selectively target molecularly defined cell classes for in vivo single-cell recordings. The targeted cells can be labeled via juxtacellular procedures and further characterized via post-hoc morphological and molecular analysis. In its current form, the protocol allows multiple recording and labeling attempts to be performed within individual animals, by means of a mechanical pipette micropositioning system. We provide proof-of-principle validation of this technique by recording from Calbindin-positive pyramidal neurons in the mouse hippocampus during spatial exploration; however, this approach can easily be extended to other behaviors and cortical or subcortical areas. The procedures described here, from the viral injection to the histological processing of brain sections, can be completed in ~4-5 weeks.This protocol is an extension to: Nat. Protoc. 9, 2369-2381 (2014): https://doi.org/10.1038/nprot.2014.161.
Collapse
Affiliation(s)
- Lingjun Ding
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Giuseppe Balsamo
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
| | - Maria Diamantaki
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Graduate Training Centre of Neuroscience-International Max-Planck Research School (IMPRS), Tübingen, Germany
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Heraklion, Greece
| | - Patricia Preston-Ferrer
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany.
| | - Andrea Burgalossi
- Institute of Neurobiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany.
| |
Collapse
|
33
|
Zhou Z, Wang X, Li X, Liao L. A bibliometric profile of optogenetics: quantitative and qualitative analyses. Front Neurosci 2023; 17:1221316. [PMID: 37424998 PMCID: PMC10323434 DOI: 10.3389/fnins.2023.1221316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Optogenetics is a rapidly developing field combining optics and genetics, with promising applications in neuroscience and beyond. However, there is currently a lack of bibliometric analyses examining publications in this area. Method Publications on optogenetics were gathered from the Web of Science Core Collection Database. A quantitative analysis was conducted to gain insights into the annual scientific output, and distribution of authors, journals, subject categories, countries, and institutions. Additionally, qualitative analysis, such as co-occurrence network analysis, thematic analysis, and theme evolution, were performed to identify the main areas and trends of optogenetics articles. Results A total of 6,824 publications were included for analysis. The number of articles has rapidly grown since 2010, with an annual growth rate of 52.82%. Deisseroth K, Boyden ES, and Hegemann P were the most prolific contributors to the field. The United States contributed the most articles (3,051 articles), followed by China (623 articles). A majority of optogenetics-related articles are published in high-quality journals, including NATURE, SCIENCE, and CELL. These articles mainly belong to four subjects: neurosciences, biochemistry and molecular biology, neuroimaging, and materials science. Co-occurrence keyword network analysis identified three clusters: optogenetic components and techniques, optogenetics and neural circuitry, optogenetics and disease. Conclusion The results suggest that optogenetics research is flourishing, focusing on optogenetic techniques and their applications in neural circuitry exploration and disease intervention. Optogenetics is expected to remain a hot topic in various fields in the future.
Collapse
Affiliation(s)
- Zhonghan Zhou
- Shandong University, Jinan, Shandong, China
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xuesheng Wang
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Xunhua Li
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Limin Liao
- Shandong University, Jinan, Shandong, China
- Department of Urology, China Rehabilitation Research Center, Beijing, China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
- China Rehabilitation Science Institute, Beijing, China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Guan S, Tian H, Yang Y, Liu M, Ding J, Wang J, Fang Y. Self-assembled ultraflexible probes for long-term neural recordings and neuromodulation. Nat Protoc 2023; 18:1712-1744. [PMID: 37248393 DOI: 10.1038/s41596-023-00824-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 02/14/2023] [Indexed: 05/31/2023]
Abstract
Ultraflexible microelectrode arrays (MEAs) that can stably record from a large number of neurons after their chronic implantation offer opportunities for understanding neural circuit mechanisms and developing next-generation brain-computer interfaces. The implementation of ultraflexible MEAs requires their reliable implantation into deep brain tissues in a minimally invasive manner, as well as their precise integration with optogenetic tools to enable the simultaneous recording of neural activity and neuromodulation. Here, we describe the process for the preparation of elastocapillary self-assembled ultraflexible MEAs, their use in combination with adeno-associated virus vectors carrying opsin genes and promoters to form an optrode probe and their in vivo experimental use in the brains of rodents, enabling electrophysiological recordings and optical modulation of neuronal activity over long periods of time (on the order of weeks to months). The procedures, including device fabrication, probe assembly and implantation, can be completed within 3 weeks. The protocol is intended to facilitate the applications of ultraflexible MEAs for long-term neuronal activity recording and combined electrophysiology and optogenetics. The protocol requires users with expertise in clean room facilities for the fabrication of ultraflexible MEAs.
Collapse
Affiliation(s)
- Shouliang Guan
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Huihui Tian
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Yinan Yang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengcheng Liu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jianfei Ding
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jinfen Wang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Ying Fang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
35
|
Rindner DJ, Lur G. Practical considerations in an era of multicolor optogenetics. Front Cell Neurosci 2023; 17:1160245. [PMID: 37293628 PMCID: PMC10244638 DOI: 10.3389/fncel.2023.1160245] [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: 02/06/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
The ability to control synaptic communication is indispensable to modern neuroscience. Until recently, only single-pathway manipulations were possible due to limited availability of opsins activated by distinct wavelengths. However, extensive protein engineering and screening efforts have drastically expanded the optogenetic toolkit, ushering in an era of multicolor approaches for studying neural circuits. Nonetheless, opsins with truly discrete spectra are scarce. Experimenters must therefore take care to avoid unintended cross-activation of optogenetic tools (crosstalk). Here, we demonstrate the multidimensional nature of crosstalk in a single model synaptic pathway, testing stimulus wavelength, irradiance, duration, and opsin choice. We then propose a "lookup table" method for maximizing the dynamic range of opsin responses on an experiment-by-experiment basis.
Collapse
Affiliation(s)
| | - Gyorgy Lur
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
36
|
Wu X, Yang F, Cai S, Pu K, Hong G. Nanotransducer-Enabled Deep-Brain Neuromodulation with NIR-II Light. ACS NANO 2023; 17:7941-7952. [PMID: 37079455 DOI: 10.1021/acsnano.2c12068] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The second near-infrared window (NIR-II window), which ranges from 1000 to 1700 nm in wavelength, exhibits distinctive advantages of reduced light scattering and thus deep penetration in biological tissues in comparison to the visible spectrum. The NIR-II window has been widely employed for deep-tissue fluorescence imaging in the past decade. More recently, deep-brain neuromodulation has been demonstrated in the NIR-II window by leveraging nanotransducers that can efficiently convert brain-penetrant NIR-II light into heat. In this Perspective, we discuss the principles and potential applications of this NIR-II deep-brain neuromodulation technique, together with its advantages and limitations compared with other existing optical methods for deep-brain neuromodulation. We also point out a few future directions where the advances in materials science and bioengineering can expand the capability and utility of NIR-II neuromodulation methods.
Collapse
Affiliation(s)
- Xiang Wu
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California 94305, USA
| | - Fan Yang
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California 94305, USA
| | - Sa Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California 94305, USA
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
| | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
37
|
Ping A, Pan L, Zhang J, Xu K, Schriver KE, Zhu J, Roe AW. Targeted Optical Neural Stimulation: A New Era for Personalized Medicine. Neuroscientist 2023; 29:202-220. [PMID: 34865559 DOI: 10.1177/10738584211057047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Targeted optical neural stimulation comprises infrared neural stimulation and optogenetics, which affect the nervous system through induced thermal transients and activation of light-sensitive proteins, respectively. The main advantage of this pair of optical tools is high functional selectivity, which conventional electrical stimulation lacks. Over the past 15 years, the mechanism, safety, and feasibility of optical stimulation techniques have undergone continuous investigation and development. When combined with other methods like optical imaging and high-field functional magnetic resonance imaging, the translation of optical stimulation to clinical practice adds high value. We review the theoretical foundations and current state of optical stimulation, with a particular focus on infrared neural stimulation as a potential bridge linking optical stimulation to personalized medicine.
Collapse
Affiliation(s)
- An Ping
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Li Pan
- Qiushi Academy for Advanced Studies (QAAS), Key Laboratory of Biomedical Engineering of Education Ministry & Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kedi Xu
- Qiushi Academy for Advanced Studies (QAAS), Key Laboratory of Biomedical Engineering of Education Ministry & Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Kenneth E Schriver
- Zhejiang University Interdisciplinary Institute of Neuroscience and Technology (ZIINT), School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junming Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Anna Wang Roe
- Zhejiang University Interdisciplinary Institute of Neuroscience and Technology (ZIINT), School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
38
|
Upconversion optogenetics-driven biohybrid sensor for infrared sensing and imaging. Acta Biomater 2023; 158:747-758. [PMID: 36638940 DOI: 10.1016/j.actbio.2023.01.017] [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: 07/22/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
Living organisms are far superior to state-of-the-art devices in visual perception as they have evolved a wide number of capabilities that encompass our most advanced technologies. By leveraging the performance of living organisms and directly interfacing them with artificial components, it can use the intricacy and metabolic efficiency of biological visual sensing within artificial machines. Inspired by the molecular basis (transient receptor potential, TRP) for infrared detection of pit-bearing organisms, we propose a TRP-like biohybrid sensor by integrating upconversion nanoparticles (UCNP) and optogenetically engineered cells on a graphene transistor for infrared sensing and imaging. The UCNP converts infrared light irradiation into blue light, the blue light activates the cells expressed with channelrhodopsin-2 (ChR2) and induces transmembrane photocurrent, and the photocurrent is detected by a biocompatible graphene transistor. Stepwise and overall experimental results show that, upon infrared light irradiation, the UCNP can rapidly mediate cellular photocurrents, which further translates into the extra output current of the graphene transistor. More notably, the response speed of the biohybrid sensor is 1∼3 orders of magnitude faster than those of TRPs heterologously expressed in cell lines in the literature, which confirms the response time advantage of the combination of UCNP and ChR2 within the sensor in place of TRPs. The biohybrid sensor can successfully image infrared targets, proving the feasibility of developing bionic infrared sensing devices by biohybrid integration of nonliving nanomaterials and biological components. This work opens up an avenue for biohybrid sensors to develop the bionic infrared vision that promisingly reproduces the functional superiority of natural organisms. STATEMENT OF SIGNIFICANCE: Infrared sensing and imaging have a wide range of military and civilian applications. Organisms have evolved excellent infrared vision with the molecular basis, transient receptor potential (TRP), and the performance is superior to existing state-of-the-art infrared devices. Inspired by this, a TRP-like biohybrid sensor based on upconversion optogenetics and a 2D material-based device is developed for infrared sensing and imaging. The biohybrid sensor has a relatively fast response speed that is 1∼3 orders of magnitude faster than that of the heterologously expressed TRPs, which enables its capability of infrared imaging with a single pixel-based method. This work broadens the spectrum of biohybrid sensing based on engineered cells to infrared, advancing the process of reproducing the excellent infrared detection of organisms.
Collapse
|
39
|
Geng Y, Li Z, Zhu J, Du C, Yuan F, Cai X, Ali A, Yang J, Tang C, Cong Z, Ma C. Advances in Optogenetics Applications for Central Nervous System Injuries. J Neurotrauma 2023. [PMID: 36305381 DOI: 10.1089/neu.2022.0290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Injuries to the central nervous system (CNS) often lead to severe neurological dysfunction and even death. However, there are still no effective measures to improve functional recovery following CNS injuries. Optogenetics, an ideal method to modulate neural activity, has shown various advantages in controlling neural circuits, promoting neural remapping, and improving cell survival. In particular, the emerging technique of optogenetics has exhibited promising therapeutic methods for CNS injuries. In this review, we introduce the light-sensitive proteins and light stimulation system that are important components of optogenetic technology in detail and summarize the development trends. In addition, we construct a comprehensive picture of the current application of optogenetics in CNS injuries and highlight recent advances for the treatment and functional recovery of neurological deficits. Finally, we discuss the therapeutic challenges and prospective uses of optogenetics therapy by photostimulation/photoinhibition modalities that would be suitable for clinical applications.
Collapse
Affiliation(s)
- Yuanming Geng
- Department of Neurosurgery, The Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenxing Li
- Department of Neurosurgery, Jinling Hospital, Nanjing, China.,Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Junhao Zhu
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chaonan Du
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feng Yuan
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiangming Cai
- School of Medicine, Southeast University, Nanjing, China
| | - Alleyar Ali
- Department of Neurosurgery, The Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Yang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chao Tang
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Zixiang Cong
- Department of Neurosurgery, Jinling Hospital, Nanjing, China
| | - Chiyuan Ma
- Department of Neurosurgery, The Affiliated Jinling Hospital of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, Nanjing, China.,Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,School of Medicine, Southeast University, Nanjing, China.,Department of Neurosurgery, Jinling Hospital, the First School of Clinical Medicine, Southern Medical University, Nanjing, China
| |
Collapse
|
40
|
Li H, Wang J, Fang Y. Recent developments in multifunctional neural probes for simultaneous neural recording and modulation. MICROSYSTEMS & NANOENGINEERING 2023; 9:4. [PMID: 36620392 PMCID: PMC9810608 DOI: 10.1038/s41378-022-00444-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/08/2022] [Accepted: 08/19/2022] [Indexed: 06/17/2023]
Abstract
Neural probes are among the most widely applied tools for studying neural circuit functions and treating neurological disorders. Given the complexity of the nervous system, it is highly desirable to monitor and modulate neural activities simultaneously at the cellular scale. In this review, we provide an overview of recent developments in multifunctional neural probes that allow simultaneous neural activity recording and modulation through different modalities, including chemical, electrical, and optical stimulation. We will focus on the material and structural design of multifunctional neural probes and their interfaces with neural tissues. Finally, future challenges and prospects of multifunctional neural probes will be discussed.
Collapse
Affiliation(s)
- Hongbian Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190 China
| | - Jinfen Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190 China
| | - Ying Fang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190 China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
41
|
Wu GY, Zheng XX, Zhao SL, Wang Y, Jiang S, Wang YS, Yi YL, Yao J, Wen HZ, Liu J, Li HL, Sui JF. The prelimbic cortex regulates itch processing by controlling attentional bias. iScience 2022; 26:105829. [PMID: 36619983 PMCID: PMC9816985 DOI: 10.1016/j.isci.2022.105829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 10/31/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Itch is a complex and unpleasant sensory experience. Recent studies have begun to investigate the neural mechanisms underlying the modulation of sensory and emotional components of itch in the brain. However, the key brain regions and neural mechanism involved in modulating the attentional processing of itch remain elusive. Here, we showed that the prelimbic cortex (PrL) is associated with itch processing and that the manipulation of itch-responsive neurons in the PrL significantly disrupted itch-induced scratching. Interestingly, we found that increasing attentional bias toward a distracting stimulus could disturb itch processing. We also demonstrated the existence of a population of attention-related neurons in the PrL that drive attentional bias to regulate itch processing. Importantly, itch-responsive neurons and attention-related neurons significantly overlapped in the PrL and were mutually interchangeable in the regulation of itch processing at the cellular activity level. Our results revealed that the PrL regulates itch processing by controlling attentional bias.
Collapse
Affiliation(s)
- Guang-Yan Wu
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Corresponding author
| | - Xiao-Xia Zheng
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Shan-Lan Zhao
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi Wang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Shan Jiang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi-Song Wang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yi-Lun Yi
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Juan Yao
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hui-Zhong Wen
- Department of Neurobiology, College of Basic Medical Sciences, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Ju Liu
- Department of Foreign Languages, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Hong-Li Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Corresponding author
| | - Jian-Feng Sui
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Department of Physiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China,Corresponding author
| |
Collapse
|
42
|
Rindner DJ, Proddutur A, Lur G. Cell-type-specific integration of feedforward and feedback synaptic inputs in the posterior parietal cortex. Neuron 2022; 110:3760-3773.e5. [PMID: 36087582 PMCID: PMC9671855 DOI: 10.1016/j.neuron.2022.08.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
The integration of feedforward (sensory) and feedback (top-down) neuronal signals is a principal function of the neocortex. Yet, we have limited insight into how these information streams are combined by individual neurons. Using a two-color optogenetic strategy, we found that layer 5 pyramidal neurons in the posterior parietal cortex receive monosynaptic dual innervation, combining sensory inputs with top-down signals. Subclasses of layer 5 pyramidal neurons integrated these synapses with distinct temporal dynamics. Specifically, regular spiking cells exhibited supralinear enhancement of delayed-but not coincident-inputs, while intrinsic burst-firing neurons selectively boosted coincident synaptic events. These subthreshold integration characteristics translated to a nonlinear summation of action potential firing. Complementing electrophysiology with computational modeling, we found that distinct integration profiles arose from a cell-type-specific interaction of ionic mechanisms and feedforward inhibition. These data provide insight into the cellular properties that guide the nonlinear interaction of distinct long-range afferents in the neocortex.
Collapse
Affiliation(s)
- Daniel J Rindner
- Department of Neurobiology and Behavior, University of California, Irvine, 1215 McGaugh Hall, Irvine, CA 92697, USA
| | - Archana Proddutur
- Department of Neurobiology and Behavior, University of California, Irvine, 1215 McGaugh Hall, Irvine, CA 92697, USA
| | - Gyorgy Lur
- Department of Neurobiology and Behavior, University of California, Irvine, 1215 McGaugh Hall, Irvine, CA 92697, USA.
| |
Collapse
|
43
|
Kathe C, Skinnider MA, Hutson TH, Regazzi N, Gautier M, Demesmaeker R, Komi S, Ceto S, James ND, Cho N, Baud L, Galan K, Matson KJE, Rowald A, Kim K, Wang R, Minassian K, Prior JO, Asboth L, Barraud Q, Lacour SP, Levine AJ, Wagner F, Bloch J, Squair JW, Courtine G. The neurons that restore walking after paralysis. Nature 2022; 611:540-547. [DOI: 10.1038/s41586-022-05385-7] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 09/23/2022] [Indexed: 11/10/2022]
Abstract
AbstractA spinal cord injury interrupts pathways from the brain and brainstem that project to the lumbar spinal cord, leading to paralysis. Here we show that spatiotemporal epidural electrical stimulation (EES) of the lumbar spinal cord1–3 applied during neurorehabilitation4,5 (EESREHAB) restored walking in nine individuals with chronic spinal cord injury. This recovery involved a reduction in neuronal activity in the lumbar spinal cord of humans during walking. We hypothesized that this unexpected reduction reflects activity-dependent selection of specific neuronal subpopulations that become essential for a patient to walk after spinal cord injury. To identify these putative neurons, we modelled the technological and therapeutic features underlying EESREHAB in mice. We applied single-nucleus RNA sequencing6–9 and spatial transcriptomics10,11 to the spinal cords of these mice to chart a spatially resolved molecular atlas of recovery from paralysis. We then employed cell type12,13 and spatial prioritization to identify the neurons involved in the recovery of walking. A single population of excitatory interneurons nested within intermediate laminae emerged. Although these neurons are not required for walking before spinal cord injury, we demonstrate that they are essential for the recovery of walking with EES following spinal cord injury. Augmenting the activity of these neurons phenocopied the recovery of walking enabled by EESREHAB, whereas ablating them prevented the recovery of walking that occurs spontaneously after moderate spinal cord injury. We thus identified a recovery-organizing neuronal subpopulation that is necessary and sufficient to regain walking after paralysis. Moreover, our methodology establishes a framework for using molecular cartography to identify the neurons that produce complex behaviours.
Collapse
|
44
|
Okuda S, Nakayama T, Uemura N, Hikawa R, Ikuno M, Yamakado H, Inoue H, Tachibana N, Hayashi Y, Takahashi R, Egawa N. Striatal-Inoculation of α-Synuclein Preformed Fibrils Aggravated the Phenotypes of REM Sleep without Atonia in A53T BAC-SNCA Transgenic Mice. Int J Mol Sci 2022; 23:13390. [PMID: 36362177 PMCID: PMC9656146 DOI: 10.3390/ijms232113390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 07/29/2023] Open
Abstract
Accumulation of α-synuclein (α-syn) is the pathological hallmark of α-synucleinopathy. Rapid eye movement (REM) sleep behavior disorder (RBD) is a pivotal manifestation of α-synucleinopathy including Parkinson's disease (PD). RBD is clinically confirmed by REM sleep without atonia (RWA) in polysomnography. To accurately characterize RWA preceding RBD and their underlying α-syn pathology, we inoculated α-syn preformed fibrils (PFFs) into the striatum of A53T human α-syn BAC transgenic (A53T BAC-SNCA Tg) mice which exhibit RBD-like phenotypes with RWA. RWA phenotypes were aggravated by PFFs-inoculation in A53T BAC-SNCA Tg mice at 1 month after inoculation, in which prominent α-syn pathology in the pedunculopontine nucleus (PPN) was observed. The intensity of RWA phenotype could be dependent on the severity of the underlying α-syn pathology.
Collapse
Affiliation(s)
- Shinya Okuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Takeo Nakayama
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Norihito Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Rie Hikawa
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Masashi Ikuno
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Haruhisa Inoue
- iPSC-Based Drug Discovery and Development Team, RIKEN BioResource Research Center, Kyoto 619-0237, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Naoko Tachibana
- Department of Neurology, Center for Sleep-Related Disorders, Kansai Electric Power Hospital, Osaka 553-0003, Japan
- Division of Sleep Medicine, Kansai Electric Power Medical Research Institute, Osaka 553-0003, Japan
| | - Yu Hayashi
- Department of Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Naohiro Egawa
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
- iPSC-Based Drug Discovery and Development Team, RIKEN BioResource Research Center, Kyoto 619-0237, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
45
|
Ko H, Yoon SP. Optogenetic neuromodulation with gamma oscillation as a new strategy for Alzheimer disease: a narrative review. JOURNAL OF YEUNGNAM MEDICAL SCIENCE 2022; 39:269-277. [PMID: 35152662 PMCID: PMC9580057 DOI: 10.12701/jyms.2021.01683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/31/2022]
Abstract
The amyloid hypothesis has been considered a major explanation of the pathogenesis of Alzheimer disease. However, failure of phase III clinical trials with anti-amyloid-beta monoclonal antibodies reveals the need for other therapeutic approaches to treat Alzheimer disease. Compared to its relatively short history, optogenetics has developed considerably. The expression of microbial opsins in cells using genetic engineering allows specific control of cell signals or molecules. The application of optogenetics to Alzheimer disease research or clinical approaches is increasing. When applied with gamma entrainment, optogenetic neuromodulation can improve Alzheimer disease symptoms. Although safety problems exist with optogenetics such as the use of viral vectors, this technique has great potential for use in Alzheimer disease. In this paper, we review the historical applications of optogenetic neuromodulation with gamma entrainment to investigate the mechanisms involved in Alzheimer disease and potential therapeutic strategies.
Collapse
Affiliation(s)
- Haneol Ko
- Medical Course, Jeju National University School of Medicine, Jeju, Korea
| | - Sang-Pil Yoon
- Department of Anatomy, Jeju National University College of Medicine, Jeju, Korea
| |
Collapse
|
46
|
Kim H, Yeow J, Najer A, Kit‐Anan W, Wang R, Rifaie‐Graham O, Thanapongpibul C, Stevens MM. Microliter Scale Synthesis of Luciferase-Encapsulated Polymersomes as Artificial Organelles for Optogenetic Modulation of Cardiomyocyte Beating. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200239. [PMID: 35901502 PMCID: PMC9507352 DOI: 10.1002/advs.202200239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/07/2022] [Indexed: 06/15/2023]
Abstract
Constructing artificial systems that effectively replace or supplement natural biological machinery within cells is one of the fundamental challenges underpinning bioengineering. At the sub-cellular scale, artificial organelles (AOs) have significant potential as long-acting biomedical implants, mimicking native organelles by conducting intracellularly compartmentalized enzymatic actions. The potency of these AOs can be heightened when judiciously combined with genetic engineering, producing highly tailorable biohybrid cellular systems. Here, the authors present a cost-effective, microliter scale (10 µL) polymersome (PSome) synthesis based on polymerization-induced self-assembly for the in situ encapsulation of Gaussia luciferase (GLuc), as a model luminescent enzyme. These GLuc-loaded PSomes present ideal features of AOs including enhanced enzymatic resistance to thermal, proteolytic, and intracellular stresses. To demonstrate their biomodulation potential, the intracellular luminescence of GLuc-loaded PSomes is coupled to optogenetically engineered cardiomyocytes, allowing modulation of cardiac beating frequency through treatment with coelenterazine (CTZ) as the substrate for GLuc. The long-term intracellular stability of the luminescent AOs allows this cardiostimulatory phenomenon to be reinitiated with fresh CTZ even after 7 days in culture. This synergistic combination of organelle-mimicking synthetic materials with genetic engineering is therefore envisioned as a highly universal strategy for the generation of new biohybrid cellular systems displaying unique triggerable properties.
Collapse
Affiliation(s)
- Hyemin Kim
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Jonathan Yeow
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Adrian Najer
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Worrapong Kit‐Anan
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Richard Wang
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Omar Rifaie‐Graham
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Chalaisorn Thanapongpibul
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| | - Molly M. Stevens
- Department of MaterialsDepartment of Bioengineeringand Institute of Biomedical EngineeringImperial College LondonLondonSW7 2AZUK
| |
Collapse
|
47
|
Amo R, Matias S, Yamanaka A, Tanaka KF, Uchida N, Watabe-Uchida M. A gradual temporal shift of dopamine responses mirrors the progression of temporal difference error in machine learning. Nat Neurosci 2022; 25:1082-1092. [PMID: 35798979 PMCID: PMC9624460 DOI: 10.1038/s41593-022-01109-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 05/24/2022] [Indexed: 02/03/2023]
Abstract
A large body of evidence has indicated that the phasic responses of midbrain dopamine neurons show a remarkable similarity to a type of teaching signal (temporal difference (TD) error) used in machine learning. However, previous studies failed to observe a key prediction of this algorithm: that when an agent associates a cue and a reward that are separated in time, the timing of dopamine signals should gradually move backward in time from the time of the reward to the time of the cue over multiple trials. Here we demonstrate that such a gradual shift occurs both at the level of dopaminergic cellular activity and dopamine release in the ventral striatum in mice. Our results establish a long-sought link between dopaminergic activity and the TD learning algorithm, providing fundamental insights into how the brain associates cues and rewards that are separated in time.
Collapse
Affiliation(s)
- Ryunosuke Amo
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Sara Matias
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Mitsuko Watabe-Uchida
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
48
|
Mondal Y, Pena RFO, Rotstein HG. Temporal filters in response to presynaptic spike trains: interplay of cellular, synaptic and short-term plasticity time scales. J Comput Neurosci 2022; 50:395-429. [DOI: 10.1007/s10827-022-00822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 10/16/2022]
|
49
|
Li H, Datunashvili M, Reyes RC, Voglmaier SM. Inositol hexakisphosphate kinases differentially regulate trafficking of vesicular glutamate transporters 1 and 2. Front Cell Neurosci 2022; 16:926794. [PMID: 35936490 PMCID: PMC9355605 DOI: 10.3389/fncel.2022.926794] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Inositol pyrophosphates have been implicated in cellular signaling and membrane trafficking, including synaptic vesicle (SV) recycling. Inositol hexakisphosphate kinases (IP6Ks) and their product, diphosphoinositol pentakisphosphate (PP-IP5 or IP7), directly and indirectly regulate proteins important in vesicle recycling by the activity-dependent bulk endocytosis pathway (ADBE). In the present study, we show that two isoforms, IP6K1 and IP6K3, are expressed in axons. The role of the kinases in SV recycling are investigated using pharmacologic inhibition, shRNA knockdown, and IP6K1 and IP6K3 knockout mice. Live-cell imaging experiments use optical reporters of SV recycling based on vesicular glutamate transporter isoforms, VGLUT1- and VGLUT2-pHluorins (pH), which recycle differently. VGLUT1-pH recycles by classical AP-2 dependent endocytosis under moderate stimulation conditions, while VGLUT2-pH recycles using AP-1 and AP-3 adaptor proteins as well. Using a short stimulus to release the readily releasable pool (RRP), we show that IP6K1 KO increases exocytosis of both VGLUT1-and VGLUT2-pH, while IP6K3 KO decreases the amount of both transporters in the RRP. In electrophysiological experiments we measure glutamate signaling with short stimuli and under the intense stimulation conditions that trigger bulk endocytosis. IP6K1 KO increases synaptic facilitation and IP6K3 KO decreases facilitation compared to wild type in CA1 hippocampal Schaffer collateral synapses. After intense stimulation, the rate of endocytosis of VGLUT2-pH, but not VGLUT1-pH, is increased by knockout, knockdown, and pharmacologic inhibition of IP6Ks. Thus IP6Ks differentially affect the endocytosis of two SV protein cargos that use different endocytic pathways. However, while IP6K1 KO and IP6K3 KO exert similar effects on endocytosis after stimulation, the isoforms exert different effects on exocytosis earlier in the stimulus and on the early phase of glutamate release. Taken together, the data indicate a role for IP6Ks both in exocytosis early in the stimulation period and in endocytosis, particularly under conditions that may utilize AP-1/3 adaptors.
Collapse
|
50
|
Sheng W, Zhao X, Huang X, Yang Y. Real-Time Image Processing Toolbox for All-Optical Closed-Loop Control of Neuronal Activities. Front Cell Neurosci 2022; 16:917713. [PMID: 35865111 PMCID: PMC9294372 DOI: 10.3389/fncel.2022.917713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
The development of in vivo imaging and optogenetic tools makes it possible to control neural circuit activities in an all-optical, closed-loop manner, but such applications are limited by the lack of software for online analysis of neuronal imaging data. We developed an analysis software ORCA (Online Real-time activity and offline Cross-session Analysis), which performs image registration, neuron segmentation, and activity extraction at over 100 frames per second, fast enough to support real-time detection and readout of neural activity. Our active neuron detection algorithm is purely statistical, achieving a much higher speed than previous methods. We demonstrated closed-loop control of neurons that were identified on the fly, without prior recording or image processing. ORCA also includes a cross-session alignment module that efficiently tracks neurons across multiple sessions. In summary, ORCA is a powerful toolbox for fast imaging data analysis and provides a solution for all-optical closed-loop control of neuronal activity.
Collapse
|