1
|
Khan WU, Shen Z, Mugo SM, Wang H, Zhang Q. Implantable hydrogels as pioneering materials for next-generation brain-computer interfaces. Chem Soc Rev 2025; 54:2832-2880. [PMID: 40035554 DOI: 10.1039/d4cs01074d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Use of brain-computer interfaces (BCIs) is rapidly becoming a transformative approach for diagnosing and treating various brain disorders. By facilitating direct communication between the brain and external devices, BCIs have the potential to revolutionize neural activity monitoring, targeted neuromodulation strategies, and the restoration of brain functions. However, BCI technology faces significant challenges in achieving long-term, stable, high-quality recordings and accurately modulating neural activity. Traditional implantable electrodes, primarily made from rigid materials like metal, silicon, and carbon, provide excellent conductivity but encounter serious issues such as foreign body rejection, neural signal attenuation, and micromotion with brain tissue. To address these limitations, hydrogels are emerging as promising candidates for BCIs, given their mechanical and chemical similarities to brain tissues. These hydrogels are particularly suitable for implantable neural electrodes due to their three-dimensional water-rich structures, soft elastomeric properties, biocompatibility, and enhanced electrochemical characteristics. These exceptional features make them ideal for signal recording, neural modulation, and effective therapies for neurological conditions. This review highlights the current advancements in implantable hydrogel electrodes, focusing on their unique properties for neural signal recording and neuromodulation technologies, with the ultimate aim of treating brain disorders. A comprehensive overview is provided to encourage future progress in this field. Implantable hydrogel electrodes for BCIs have enormous potential to influence the broader scientific landscape and drive groundbreaking innovations across various sectors.
Collapse
Affiliation(s)
- Wasid Ullah Khan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Zhenzhen Shen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Samuel M Mugo
- Department of Physical Sciences, MacEwan University, Edmonton, ABT5J4S2, Canada
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Qiang Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
- CAS Applied Chemistry Science & Technology Co., Ltd, Changchun, Jilin 130022, P. R. China
| |
Collapse
|
2
|
Yang X, Kubican SE, Yi Z, Tong S. Advances in magnetic nanoparticles for molecular medicine. Chem Commun (Camb) 2025; 61:3093-3108. [PMID: 39846549 PMCID: PMC11756346 DOI: 10.1039/d4cc05167j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Magnetic nanoparticles (MNPs) are highly versatile nanomaterials in nanomedicine, owing to their diverse magnetic properties, which can be tailored through variations in size, shape, composition, and exposure to inductive magnetic fields. Over four decades of research have led to the clinical approval or ongoing trials of several MNP formulations, fueling continued innovation. Beyond traditional applications in drug delivery, imaging, and cancer hyperthermia, MNPs have increasingly advanced into molecular medicine. Under external magnetic fields, MNPs can generate mechano- or thermal stimuli to modulate individual molecules or cells deep within tissue, offering precise, remote control of biological processes at cellular and molecular levels. These unique capabilities have opened new avenues in emerging fields such as genome editing, cell therapies, and neuroscience, underpinned by a growing understanding of nanomagnetism and the molecular mechanisms responding to mechanical and thermal cues. Research on MNPs as a versatile synthetic material capable of engineering control at the cellular and molecular levels holds great promise for advancing the frontiers of molecular medicine, including areas such as genome editing and synthetic biology. This review summarizes recent clinical studies showcasing the classical applications of MNPs and explores their integration into molecular medicine, with the goal of inspiring the development of next-generation MNP-based platforms for disease treatment.
Collapse
Affiliation(s)
- Xiaoyue Yang
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Sarah E Kubican
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Zhongchao Yi
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| | - Sheng Tong
- F. Joseph Halcomb III, M. D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky 40536, USA.
| |
Collapse
|
3
|
Wang W, Shi Y, Chai W, Tang KWK, Pyatnitskiy I, Xie Y, Liu X, He W, Jeong J, Hsieh JC, Lozano AR, Artman B, Shi X, Hoefer N, Shrestha B, Stern NB, Zhou W, McComb DW, Porter T, Henkelman G, Chen B, Wang H. H-bonded organic frameworks as ultrasound-programmable delivery platform. Nature 2025; 638:401-410. [PMID: 39910310 PMCID: PMC12038167 DOI: 10.1038/s41586-024-08401-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 11/14/2024] [Indexed: 02/07/2025]
Abstract
The precise control of mechanochemical activation within deep tissues using non-invasive ultrasound holds profound implications for advancing our understanding of fundamental biomedical sciences and revolutionizing disease treatments1-4. However, a theory-guided mechanoresponsive materials system with well-defined ultrasound activation has yet to be explored5,6. Here we present the concept of using porous hydrogen-bonded organic frameworks (HOFs) as toolkits for focused ultrasound (FUS) programmably triggered drug activation to control specific cellular events in the deep brain, through on-demand scission of the supramolecular interactions. A theoretical model is developed to potentially visualize the mechanochemical scission and ultrasound mechanics, providing valuable guidelines for the rational design of mechanoresponsive materials to achieve programmable control. To demonstrate the practicality of this approach, we encapsulate the designer drug clozapine N-oxide (CNO) into the optimal HOF nanocrystals for FUS-gated release to activate engineered G-protein-coupled receptors in the ventral tegmental area (VTA) of mice and rats and hence achieve targeted neural circuit modulation even at depth 9 mm with a latency of seconds. This work demonstrates the capability of ultrasound to precisely control molecular interactions and develops ultrasound-programmable HOFs to non-invasively and spatiotemporally control cellular events, thereby facilitating the establishment of precise molecular therapeutic possibilities.
Collapse
Affiliation(s)
- Wenliang Wang
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Yanshu Shi
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Wenrui Chai
- Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Kai Wing Kevin Tang
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Ilya Pyatnitskiy
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Yi Xie
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Xiangping Liu
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Weilong He
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Jinmo Jeong
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Ju-Chun Hsieh
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Anakaren Romero Lozano
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Brinkley Artman
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Xi Shi
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Nicole Hoefer
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - Binita Shrestha
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Noah B Stern
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Wei Zhou
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - Tyrone Porter
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Graeme Henkelman
- Freshman Research Initiative, College of Natural Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Banglin Chen
- Department of Chemistry, The University of Texas at San Antonio, San Antonio, TX, USA.
| | - Huiliang Wang
- Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
4
|
Pan Y, Pan C, Mao L, Yu P. Neuromodulation with chemicals: Opportunities and challenges. FUNDAMENTAL RESEARCH 2025; 5:55-62. [PMID: 40166084 PMCID: PMC11955035 DOI: 10.1016/j.fmre.2024.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/02/2025] Open
Abstract
Chemicals play a crucial role in neurophysiological and neuropathological processes. By regulating the concentration of specific chemicals, receptors on the neuron cell membrane can be modulated to activate or inhibit, thereby influencing specific ion channels and facilitating neuromodulation. This review introduces several chemical modulation techniques, such as microinjection, electrode/nanoparticle-based chemical delivery methods, in situ electrochemical synthesis and chemogenetics. While these techniques show promise in expanding the application of chemical neuromodulation, they currently exhibit different degrees of shortcomings and room for improvement. This review summarizes the opportunities and challenges for chemical neuromodulation methods and provide an outlook for their prospects in the future.
Collapse
Affiliation(s)
- Yifei Pan
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Cong Pan
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Lanqun Mao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ping Yu
- Key Laboratory of Analytical Chemistry for Living Biosystems Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
5
|
Kim YJ, Kent N, Vargas Paniagua E, Driscoll N, Tabet A, Koehler F, Malkin E, Frey E, Manthey M, Sahasrabudhe A, Cannon TM, Nagao K, Mankus D, Bisher M, de Nola G, Lytton-Jean A, Signorelli L, Gregurec D, Anikeeva P. Magnetoelectric nanodiscs enable wireless transgene-free neuromodulation. NATURE NANOTECHNOLOGY 2025; 20:121-131. [PMID: 39394431 PMCID: PMC11750723 DOI: 10.1038/s41565-024-01798-9] [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: 01/17/2024] [Accepted: 09/02/2024] [Indexed: 10/13/2024]
Abstract
Deep brain stimulation with implanted electrodes has transformed neuroscience studies and treatment of neurological and psychiatric conditions. Discovering less invasive alternatives to deep brain stimulation could expand its clinical and research applications. Nanomaterial-mediated transduction of magnetic fields into electric potentials has been explored as a means for remote neuromodulation. Here we synthesize magnetoelectric nanodiscs (MENDs) with a core-double-shell Fe3O4-CoFe2O4-BaTiO3 architecture (250 nm diameter and 50 nm thickness) with efficient magnetoelectric coupling. We find robust responses to magnetic field stimulation in neurons decorated with MENDs at a density of 1 µg mm-2 despite individual-particle potentials below the neuronal excitation threshold. We propose a model for repetitive subthreshold depolarization that, combined with cable theory, supports our observations in vitro and informs magnetoelectric stimulation in vivo. Injected into the ventral tegmental area or the subthalamic nucleus of genetically intact mice at concentrations of 1 mg ml-1, MENDs enable remote control of reward or motor behaviours, respectively. These findings set the stage for mechanistic optimization of magnetoelectric neuromodulation towards applications in neuroscience research.
Collapse
Affiliation(s)
- Ye Ji Kim
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Noah Kent
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emmanuel Vargas Paniagua
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicolette Driscoll
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florian Koehler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elian Malkin
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department Engineering in Computation and Cognition, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ethan Frey
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie Manthey
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Atharva Sahasrabudhe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Taylor M Cannon
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Keisuke Nagao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David Mankus
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Margaret Bisher
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni de Nola
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Abigail Lytton-Jean
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lorenzo Signorelli
- Department of Chemistry and Pharmacy, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Danijela Gregurec
- Department of Chemistry and Pharmacy, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Hou Z. The New Era of Neural Modulation Led by Smart Nanomaterials. Int J Nanomedicine 2024; 19:12287-12295. [PMID: 39588257 PMCID: PMC11586479 DOI: 10.2147/ijn.s491440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024] Open
Abstract
Understanding the physiology and pathology of neural circuits is crucial in neuroscience research. A variety of techniques have been utilized in medical research, with several established methods applied in clinical therapy to enhance patient' neurological functions. Traditional methods include generating electric fields near neural tissue using electrodes, or non-contact modulation using light, chemicals, magnetic fields, and ultrasound. The advent of nanotechnology represents a new advancement in neural modulation techniques, offering high precision and the ability to target specific cell types. Smart nanomaterials enable the conversion of remote signals (such as light, magnetic, or ultrasound) into local stimuli (eg, electric fields or heat) for neurons. Surface treatment technologies of nanomaterials have enhanced biocompatibility, making targeted delivery to specific cell types possible and paving the way for precise neural modulation. This perspective will explore neural modulation techniques supported by nanomedical materials, focusing on photoelectric, photothermal, magnetoelectric, magnetothermal, and acoustoelectric conversion mechanisms, and looking forward to their medical applications.
Collapse
Affiliation(s)
- Zhitao Hou
- College of Basic Medical and Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, People’s Republic of China
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Neurology, the Second Hospital Affiliated with Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150010, People’s Republic of China
- Department of Neurology, the First Hospital Affiliated with Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150010, People’s Republic of China
- Institute of Shaanxi Key Laboratory of Ultrasonics, Shaanxi Normal University, Xi’an, 710119, People’s Republic of China
| |
Collapse
|
7
|
Cai AJ, Gao K, Zhang F, Jiang YW. Recent advances and current status of gene therapy for epilepsy. World J Pediatr 2024; 20:1115-1137. [PMID: 39395088 DOI: 10.1007/s12519-024-00843-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/05/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Epilepsy is a common neurological disorder with complex pathogenic mechanisms, and refractory epilepsy often lacks effective treatments. Gene therapy is a promising therapeutic option, with various preclinical experiments achieving positive results, some of which have progressed to clinical studies. DATA SOURCES This narrative review was conducted by searching for papers published in PubMed/MEDLINE with the following single and/or combination keywords: epilepsy, children, neurodevelopmental disorders, genetics, gene therapy, vectors, transgenes, receptors, ion channels, micro RNAs (miRNAs), clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (Cas)9 (CRISPR/Cas9), expression regulation, optogenetics, chemical genetics, mitochondrial epilepsy, challenges, ethics, and disease models. RESULTS Currently, gene therapy research in epilepsy primarily focuses on symptoms attenuation mediated by viral vectors such as adeno-associated virus and other types. Advances in gene therapy technologies, such as CRISPR/Cas9, have provided a new direction for epilepsy treatment. However, the clinical application still faces several challenges, including issues related to vectors, models, expression controllability, and ethical considerations. CONCLUSIONS Here, we summarize the relevant research and clinical advances in gene therapy for epilepsy and outline the challenges facing its clinical application. In addition to the shortcomings inherent in gene therapy components, the reconfiguration of excitatory and inhibitory properties in epilepsy treatment is a delicate process. On-demand, cell-autonomous treatments and multidisciplinary collaborations may be crucial in addressing these issues. Understanding gene therapy for epilepsy will help clinicians gain a clearer perception of the research progress and challenges, guiding the design of future clinical protocols and research decisions.
Collapse
Affiliation(s)
- Ao-Jie Cai
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Henan Province, Zhengzhou, 450052, China
| | - Kai Gao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Fan Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China
- Children Epilepsy Center, Peking University First Hospital, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China.
- Beijing Key Laboratory of Molecular Diagnosis and Study on Pediatric Genetic Diseases, Beijing, China.
- Children Epilepsy Center, Peking University First Hospital, Beijing, China.
- Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
8
|
Matuwana D, Hong E, Huang S, Xu X, Jang G, Xiao R, Rao S, Wang Q. Near-infrared activated liposomes for neuroprotection in glaucoma. J Mater Chem B 2024; 12:10902-10914. [PMID: 39355895 DOI: 10.1039/d4tb00745j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Neurodegenerative diseases have a profound impact on vision, leading to conditions such as glaucoma, optic neuropathy, and diabetic retinopathy, affecting millions worldwide. These diseases are characterized by the degeneration of retinal ganglion cells (RGCs), resulting in a progressive loss of visual acuity and field, with the threat of irreversible blindness. However, existing treatments, such as eye drops, direct injections, and laser surgeries face significant challenges due to limited efficacy and potential infection. The inefficiency of traditional corneal drug delivery methods is a major obstacle in treating vision neurodegenerative diseases. To address these challenges, we developed a remotely triggered on-demand liposomal delivery system to treat glaucomatous neurodegeneration in mice. We utilized the localized surface plasmon resonance (LSPR) effect of gold nanorods (AuNRs) under near-infrared (NIR) light (808 nm) to control the release of cyclodextrin-encapsulated melatonin from thermally responsive liposomal nanocarriers in the vitreous humor. Due to the transparency of the eye's cornea, NIR light can penetrate deep tissues, enabling on-demand drug delivery to the retina. By enhancing the drug's solubility and stability through cyclodextrin encapsulation, this remotely activated melatonin/HPβCD AuNRs liposomes delivery system can decrease intraocular pressure (IOP) elevation by (24 ± 7)%, enhance the survival rate of RGCs by (77 ± 6)%, and decrease glial fibrillary acidic protein (GFAP) activation by (75 ± 6)% at depth in an acute experimental glaucoma model. This NIR-triggered drug delivery system presents the potential of a promising minimally photo-triggered therapeutic option for glaucoma treatment.
Collapse
Affiliation(s)
- Dorcas Matuwana
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| | - Eunji Hong
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| | - Sizhe Huang
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| | - Xinxin Xu
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| | - Geunho Jang
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| | - Ruobai Xiao
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| | - Siyuan Rao
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
- Integrative Neuroscience Program, Binghamton University, Binghamton, NY 13902, USA
| | - Qianbin Wang
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY 13902, USA.
| |
Collapse
|
9
|
Liu S, Sun J. Magnetic nanomaterials mediate precise magnetic therapy. Biomed Phys Eng Express 2024; 10:052001. [PMID: 38981447 DOI: 10.1088/2057-1976/ad60cb] [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: 01/23/2024] [Accepted: 07/09/2024] [Indexed: 07/11/2024]
Abstract
Magnetic nanoparticle (MNP)-mediated precision magnet therapy plays a crucial role in treating various diseases. This therapeutic strategy compensates for the limitations of low spatial resolution and low focusing of magnetic stimulation, and realizes the goal of wireless teletherapy with precise targeting of focal areas. This paper summarizes the preparation methods of magnetic nanomaterials, the properties of magnetic nanoparticles, the biological effects, and the measurement methods for detecting magnetism; discusses the research progress of precision magnetotherapy in the treatment of psychiatric disorders, neurological injuries, metabolic disorders, and bone-related disorders, and looks forward to the future development trend of precision magnet therapy.
Collapse
Affiliation(s)
- Sha Liu
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| | - Jianfei Sun
- Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, People's Republic of China
| |
Collapse
|
10
|
Quelhas KN, Henn MA, Farias R, Tew WL, Woods SI. GPU-accelerated parallel image reconstruction strategies for magnetic particle imaging. Phys Med Biol 2024; 69:135005. [PMID: 38843809 DOI: 10.1088/1361-6560/ad5510] [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: 12/26/2023] [Accepted: 06/06/2024] [Indexed: 06/25/2024]
Abstract
Objective. Image reconstruction is a fundamental step in magnetic particle imaging (MPI). One of the main challenges is the fact that the reconstructions are computationally intensive and time-consuming, so choosing an algorithm presents a compromise between accuracy and execution time, which depends on the application. This work proposes a method that provides both fast and accurate image reconstructions.Approach. Image reconstruction algorithms were implemented to be executed in parallel ingraphics processing units(GPUs) using the CUDA framework. The calculation of the model-based MPI calibration matrix was also implemented in GPU to allow both fast and flexible reconstructions.Main results. The parallel algorithms were able to accelerate the reconstructions by up to about6,100times in comparison to the serial Kaczmarz algorithm executed in the CPU, allowing for real-time applications. Reconstructions using the OpenMPIData dataset validated the proposed algorithms and demonstrated that they are able to provide both fast and accurate reconstructions. The calculation of the calibration matrix was accelerated by up to about 37 times.Significance. The parallel algorithms proposed in this work can provide single-frame MPI reconstructions in real time, with frame rates greater than 100 frames per second. The parallel calculation of the calibration matrix can be combined with the parallel reconstruction to deliver images in less time than the serial Kaczmarz reconstruction, potentially eliminating the need of storing the calibration matrix in the main memory, and providing the flexibility of redefining scanning and reconstruction parameters during execution.
Collapse
Affiliation(s)
- Klaus N Quelhas
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, United States of America
- Systems Engineering and Computer Science Program, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mark-Alexander Henn
- Information Technology Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, United States of America
- University of Maryland, College Park, MD, United States of America
| | - Ricardo Farias
- Systems Engineering and Computer Science Program, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Weston L Tew
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, United States of America
| | - Solomon I Woods
- Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD, United States of America
| |
Collapse
|
11
|
Shi J, Tan C, Ge X, Qin Z, Xiong H. Recent advances in stimuli-responsive controlled release systems for neuromodulation. J Mater Chem B 2024; 12:5769-5786. [PMID: 38804184 DOI: 10.1039/d4tb00720d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Neuromodulation aims to modulate the signaling activity of neurons or neural networks by the precise delivery of electrical stimuli or chemical agents and is crucial for understanding brain function and treating brain disorders. Conventional approaches, such as direct physical stimulation through electrical or acoustic methods, confront challenges stemming from their invasive nature, dependency on wired power sources, and unstable therapeutic outcomes. The emergence of stimulus-responsive delivery systems harbors the potential to revolutionize neuromodulation strategies through the precise and controlled release of neurochemicals in a specific brain region. This review comprehensively examines the biological barriers controlled release systems may encounter in vivo and the recent advances and applications of these systems in neuromodulation. We elucidate the intricate interplay between the molecular structure of delivery systems and response mechanisms to furnish insights for material selection and design. Additionally, the review contemplates the prospects and challenges associated with these systems in neuromodulation. The overarching objective is to propel the application of neuromodulation technology in analyzing brain functions, treating brain disorders, and providing insightful perspectives for exploiting new systems for biomedical applications.
Collapse
Affiliation(s)
- Jielin Shi
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Chao Tan
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Xiaoqian Ge
- Department of Mechanical Engineering, The University of Texas at Dallas Richardson, TX 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas Richardson, TX 75080, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Hejian Xiong
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Huang S, Liu X, Lin S, Glynn C, Felix K, Sahasrabudhe A, Maley C, Xu J, Chen W, Hong E, Crosby AJ, Wang Q, Rao S. Control of polymers' amorphous-crystalline transition enables miniaturization and multifunctional integration for hydrogel bioelectronics. Nat Commun 2024; 15:3525. [PMID: 38664445 PMCID: PMC11045824 DOI: 10.1038/s41467-024-47988-w] [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: 04/26/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Soft bioelectronic devices exhibit motion-adaptive properties for neural interfaces to investigate complex neural circuits. Here, we develop a fabrication approach through the control of metamorphic polymers' amorphous-crystalline transition to miniaturize and integrate multiple components into hydrogel bioelectronics. We attain an about 80% diameter reduction in chemically cross-linked polyvinyl alcohol hydrogel fibers in a fully hydrated state. This strategy allows regulation of hydrogel properties, including refractive index (1.37-1.40 at 480 nm), light transmission (>96%), stretchability (139-169%), bending stiffness (4.6 ± 1.4 N/m), and elastic modulus (2.8-9.3 MPa). To exploit the applications, we apply step-index hydrogel optical probes in the mouse ventral tegmental area, coupled with fiber photometry recordings and social behavioral assays. Additionally, we fabricate carbon nanotubes-PVA hydrogel microelectrodes by incorporating conductive nanomaterials in hydrogel for spontaneous neural activities recording. We enable simultaneous optogenetic stimulation and electrophysiological recordings of light-triggered neural activities in Channelrhodopsin-2 transgenic mice.
Collapse
Affiliation(s)
- Sizhe Huang
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Xinyue Liu
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - Shaoting Lin
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - Christopher Glynn
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Kayla Felix
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Atharva Sahasrabudhe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Collin Maley
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Jingyi Xu
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Weixuan Chen
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Eunji Hong
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Alfred J Crosby
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA, USA
| | - Qianbin Wang
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA.
| | - Siyuan Rao
- Department of Biomedical Engineering, Binghamton University, State University of New York, Binghamton, NY, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
13
|
Xie X, Zhai J, Zhou X, Guo Z, Lo PC, Zhu G, Chan KWY, Yang M. Magnetic Particle Imaging: From Tracer Design to Biomedical Applications in Vasculature Abnormality. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306450. [PMID: 37812831 DOI: 10.1002/adma.202306450] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/14/2023] [Indexed: 10/11/2023]
Abstract
Magnetic particle imaging (MPI) is an emerging non-invasive tomographic technique based on the response of magnetic nanoparticles (MNPs) to oscillating drive fields at the center of a static magnetic gradient. In contrast to magnetic resonance imaging (MRI), which is driven by uniform magnetic fields and projects the anatomic information of the subjects, MPI directly tracks and quantifies MNPs in vivo without background signals. Moreover, it does not require radioactive tracers and has no limitations on imaging depth. This article first introduces the basic principles of MPI and important features of MNPs for imaging sensitivity, spatial resolution, and targeted biodistribution. The latest research aiming to optimize the performance of MPI tracers is reviewed based on their material composition, physical properties, and surface modifications. While the unique advantages of MPI have led to a series of promising biomedical applications, recent development of MPI in investigating vascular abnormalities in cardiovascular and cerebrovascular systems, and cancer are also discussed. Finally, recent progress and challenges in the clinical translation of MPI are discussed to provide possible directions for future research and development.
Collapse
Affiliation(s)
- Xulin Xie
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Jiao Zhai
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Zhengjun Guo
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
- Department of Oncology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Pui-Chi Lo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Kannie W Y Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, China
| |
Collapse
|
14
|
Stoddart PR, Begeng JM, Tong W, Ibbotson MR, Kameneva T. Nanoparticle-based optical interfaces for retinal neuromodulation: a review. Front Cell Neurosci 2024; 18:1360870. [PMID: 38572073 PMCID: PMC10987880 DOI: 10.3389/fncel.2024.1360870] [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: 12/24/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Degeneration of photoreceptors in the retina is a leading cause of blindness, but commonly leaves the retinal ganglion cells (RGCs) and/or bipolar cells extant. Consequently, these cells are an attractive target for the invasive electrical implants colloquially known as "bionic eyes." However, after more than two decades of concerted effort, interfaces based on conventional electrical stimulation approaches have delivered limited efficacy, primarily due to the current spread in retinal tissue, which precludes high-acuity vision. The ideal prosthetic solution would be less invasive, provide single-cell resolution and an ability to differentiate between different cell types. Nanoparticle-mediated approaches can address some of these requirements, with particular attention being directed at light-sensitive nanoparticles that can be accessed via the intrinsic optics of the eye. Here we survey the available known nanoparticle-based optical transduction mechanisms that can be exploited for neuromodulation. We review the rapid progress in the field, together with outstanding challenges that must be addressed to translate these techniques to clinical practice. In particular, successful translation will likely require efficient delivery of nanoparticles to stable and precisely defined locations in the retinal tissues. Therefore, we also emphasize the current literature relating to the pharmacokinetics of nanoparticles in the eye. While considerable challenges remain to be overcome, progress to date shows great potential for nanoparticle-based interfaces to revolutionize the field of visual prostheses.
Collapse
Affiliation(s)
- Paul R. Stoddart
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - James M. Begeng
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Wei Tong
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- School of Physics, The University of Melbourne, Melbourne, VIC, Australia
| | - Michael R. Ibbotson
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Tatiana Kameneva
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC, Australia
| |
Collapse
|
15
|
Chen JC, Bhave G, Alrashdan F, Dhuliyawalla A, Hogan KJ, Mikos AG, Robinson JT. Self-rectifying magnetoelectric metamaterials for remote neural stimulation and motor function restoration. NATURE MATERIALS 2024; 23:139-146. [PMID: 37814117 PMCID: PMC10972531 DOI: 10.1038/s41563-023-01680-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 09/04/2023] [Indexed: 10/11/2023]
Abstract
Magnetoelectric materials convert magnetic fields into electric fields. These materials are often used in wireless electronic and biomedical applications. For example, magnetoelectrics could enable the remote stimulation of neural tissue, but the optimal resonance frequencies are typically too high to stimulate neural activity. Here we describe a self-rectifying magnetoelectric metamaterial for a precisely timed neural stimulation. This metamaterial relies on nonlinear charge transport across semiconductor layers that allow the material to generate a steady bias voltage in the presence of an alternating magnetic field. We generate arbitrary pulse sequences with time-averaged voltage biases in excess of 2 V. As a result, we can use magnetoelectric nonlinear metamaterials to wirelessly stimulate peripheral nerves to restore a sensory reflex in an anaesthetized rat model and restore signal propagation in a severed nerve with latencies of less than 5 ms. Overall, these results showing the rational design of magnetoelectric metamaterials support applications in advanced biotechnology and electronics.
Collapse
Affiliation(s)
- Joshua C Chen
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Gauri Bhave
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Fatima Alrashdan
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Abdeali Dhuliyawalla
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Katie J Hogan
- Department of Bioengineering, Rice University, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | | | - Jacob T Robinson
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA.
- Applied Physics Program, Rice University, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
16
|
Wang W, Kevin Tang KW, Pyatnitskiy I, Liu X, Shi X, Huo D, Jeong J, Wynn T, Sangani A, Baker A, Hsieh JC, Lozano AR, Artman B, Fenno L, Buch VP, Wang H. Ultrasound-Induced Cascade Amplification in a Mechanoluminescent Nanotransducer for Enhanced Sono-Optogenetic Deep Brain Stimulation. ACS NANO 2023; 17:24936-24946. [PMID: 38096422 PMCID: PMC10932741 DOI: 10.1021/acsnano.3c06577] [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] [Indexed: 12/27/2023]
Abstract
Remote and genetically targeted neuromodulation in the deep brain is important for understanding and treatment of neurological diseases. Ultrasound-triggered mechanoluminescent technology offers a promising approach for achieving remote and genetically targeted brain modulation. However, its application has thus far been limited to shallow brain depths due to challenges related to low sonochemical reaction efficiency and restricted photon yields. Here we report a cascaded mechanoluminescent nanotransducer to achieve efficient light emission upon ultrasound stimulation. As a result, blue light was generated under ultrasound stimulation with a subsecond response latency. Leveraging the high energy transfer efficiency of focused ultrasound in brain tissue and the high sensitivity to ultrasound of these mechanoluminescent nanotransducers, we are able to show efficient photon delivery and activation of ChR2-expressing neurons in both the superficial motor cortex and deep ventral tegmental area after intracranial injection. Our liposome nanotransducers enable minimally invasive deep brain stimulation for behavioral control in animals via a flexible, mechanoluminescent sono-optogenetic system.
Collapse
Affiliation(s)
- Wenliang Wang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kai Wing Kevin Tang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ilya Pyatnitskiy
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Xiangping Liu
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Xi Shi
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas 78712, United States
| | - David Huo
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jinmo Jeong
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Thomas Wynn
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Arjun Sangani
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Andrew Baker
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ju-Chun Hsieh
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Anakaren Romero Lozano
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Brinkley Artman
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Lief Fenno
- Department of Psychiatry & Behavioral Science, The University of Texas at Austin Dell Medical School, Austin, Texas 78712, United States
| | - Vivek P Buch
- Department of Neurosurgery, Stanford University, Stanford, California 94304, United States
| | - Huiliang Wang
- Biomedical Engineering Cockrell School of Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
17
|
Kim YJ, Driscoll N, Kent N, Paniagua EV, Tabet A, Koehler F, Manthey M, Sahasrabudhe A, Signorelli L, Gregureć D, Anikeeva P. Magnetoelectric Nanodiscs Enable Wireless Transgene-Free Neuromodulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.24.573272. [PMID: 38234742 PMCID: PMC10793401 DOI: 10.1101/2023.12.24.573272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Deep-brain stimulation (DBS) with implanted electrodes revolutionized treatment of movement disorders and empowered neuroscience studies. Identifying less invasive alternatives to DBS may further extend its clinical and research applications. Nanomaterial-mediated transduction of magnetic fields into electric potentials offers an alternative to invasive DBS. Here, we synthesize magnetoelectric nanodiscs (MENDs) with a core-double shell Fe3O4-CoFe2O4-BaTiO3 architecture with efficient magnetoelectric coupling. We find robust responses to magnetic field stimulation in neurons decorated with MENDs at a density of 1 μg/mm2 despite individual-particle potentials below the neuronal excitation threshold. We propose a model for repetitive subthreshold depolarization, which combined with cable theory, corroborates our findings in vitro and informs magnetoelectric stimulation in vivo. MENDs injected into the ventral tegmental area of genetically intact mice at concentrations of 1 mg/mL enable remote control of reward behavior, setting the stage for mechanistic optimization of magnetoelectric neuromodulation and inspiring its future applications in fundamental and translational neuroscience.
Collapse
Affiliation(s)
- Ye Ji Kim
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicolette Driscoll
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Noah Kent
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emmanuel Vargas Paniagua
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anthony Tabet
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florian Koehler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie Manthey
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Atharva Sahasrabudhe
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lorenzo Signorelli
- Department of Chemistry and Pharmacy, Friedrich-Alexander University of Erlangen - Nuremberg, Erlangen, Germany
| | - Danijela Gregureć
- Department of Chemistry and Pharmacy, Friedrich-Alexander University of Erlangen - Nuremberg, Erlangen, Germany
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
18
|
Wang W, Shi Y, Chai W, Kevin Tang KW, Pyatnitskiy I, Xie Y, Liu X, He W, Jeong J, Hsieh JC, Lozano AR, Artman B, Henkelman G, Chen B, Wang H. Ultrasound programmable hydrogen-bonded organic frameworks for sono-chemogenetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570721. [PMID: 38106007 PMCID: PMC10723392 DOI: 10.1101/2023.12.08.570721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
The precise control of mechanochemical activation within deep tissues via non-invasive ultrasound holds profound implications for advancing our understanding of fundamental biomedical sciences and revolutionizing disease treatments. However, a theory-guided mechanoresponsive materials system with well-defined ultrasound activation has yet to be explored. Here we present the concept of using porous hydrogen-bonded organic frameworks (HOFs) as toolkits for focused ultrasound programmably triggered drug activation to control specific cellular events in the deep brain, through on-demand scission of the supramolecular interactions. A theoretical model is developed to visualize the mechanochemical scission and ultrasound mechanics, providing valuable guidelines for the rational design of mechanoresponsive materials at the molecular level to achieve programmable and spatiotemporal activation control. To demonstrate the practicality of this approach, we encapsulate designer drug clozapine N-oxide (CNO) into the optimal HOF nanoparticles for FUS gated release to activate engineered G-protein-coupled receptors in the mice and rat ventral tegmental area (VTA), and hence achieved targeted neural circuits modulation even at depth 9 mm with a latency of seconds. This work demonstrates the capability of ultrasound to precisely control molecular interaction and develops ultrasound programmable HOFs to minimally invasive and spatiotemporally control cellular events, thereby facilitating the establishment of precise molecular therapeutic possibilities. We anticipate that this research could serve as a source of inspiration for precise and non-invasive molecular manipulation techniques, potentially applicable in programming molecular robots to achieve sophisticated control over cellular events in deep tissues.
Collapse
|
19
|
Berndt A, Cai D, Cohen A, Juarez B, Iglesias JT, Xiong H, Qin Z, Tian L, Slesinger PA. Current Status and Future Strategies for Advancing Functional Circuit Mapping In Vivo. J Neurosci 2023; 43:7587-7598. [PMID: 37940594 PMCID: PMC10634581 DOI: 10.1523/jneurosci.1391-23.2023] [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: 07/21/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 11/10/2023] Open
Abstract
The human brain represents one of the most complex biological systems, containing billions of neurons interconnected through trillions of synapses. Inherent to the brain is a biochemical complexity involving ions, signaling molecules, and peptides that regulate neuronal activity and allow for short- and long-term adaptations. Large-scale and noninvasive imaging techniques, such as fMRI and EEG, have highlighted brain regions involved in specific functions and visualized connections between different brain areas. A major shortcoming, however, is the need for more information on specific cell types and neurotransmitters involved, as well as poor spatial and temporal resolution. Recent technologies have been advanced for neuronal circuit mapping and implemented in behaving model organisms to address this. Here, we highlight strategies for targeting specific neuronal subtypes, identifying, and releasing signaling molecules, controlling gene expression, and monitoring neuronal circuits in real-time in vivo Combined, these approaches allow us to establish direct causal links from genes and molecules to the systems level and ultimately to cognitive processes.
Collapse
Affiliation(s)
| | - Denise Cai
- Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | | | | | | | - Zhenpeng Qin
- University of Texas-Dallas, Richardson, TX 75080
| | - Lin Tian
- University of California-Davis, Davis, CA 95616
| | | |
Collapse
|
20
|
Li W, Huberman-Shlaes J, Tian B. Perspectives on Multiscale Colloid-Based Materials for Biomedical Applications. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:13759-13769. [PMID: 37733490 PMCID: PMC10552542 DOI: 10.1021/acs.langmuir.3c01274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Indexed: 09/23/2023]
Abstract
Colloid-based materials with tunable biophysical and chemical properties have demonstrated significant potential in a wide range of biomedical applications. The ability to manipulate these properties across various size scales, encompassing nano-, micro-, and macrodomains, is essential to enhancing current biomedical technologies and facilitating the development of novel applications. Focusing on material design, we explore various synthetic colloid-based materials at the nano- and microscales and investigate their correlation with biological systems. Furthermore, we examine the utilization of the self-assembly of colloids to construct monolithic and macroscopic materials suitable for biointerfaces. By probing the potential of spatial imaging and localized drug delivery, enhanced functionality, and colloidal manipulation, we highlight emerging opportunities that could significantly advance the field of colloid-based materials in biomedical applications.
Collapse
Affiliation(s)
- Wen Li
- Department
of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Judah Huberman-Shlaes
- Department
of Biology, The University of Chicago, Chicago, Illinois 60637, United States
| | - Bozhi Tian
- Department
of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
- The
James Franck Institute, The University of
Chicago, Chicago, Illinois 60637, United States
- The
Institute for Biophysical Dynamics, The
University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
21
|
Huang S, Villafranca SU, Mehta I, Yosfan O, Hong E, Wang A, Wu N, Wang Q, Rao S. A nanoscale inorganic coating strategy for stabilizing hydrogel neural probes in vivo. J Mater Chem B 2023; 11:7629-7640. [PMID: 37401386 PMCID: PMC10530439 DOI: 10.1039/d3tb00710c] [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] [Indexed: 07/05/2023]
Abstract
Hydrogels with adaptable optical and mechanical characteristics show considerable promise for light delivery in vivo with neuroengineering applications. However, the unlinked amorphous polymer chains within hydrogels can cause volumetric swelling after water absorption under physiological conditions over time. Chemically cross-linked poly(vinyl alcohol) (PVA) hydrogels showcase fatigue-resistant attributes and promising biocompatibility for the manufacture of soft neural probes. However, possible swelling of the PVA hydrogel matrix could impact the structural stability of hydrogel-based bioelectronics and their long-term in vivo functionality. In this study, we utilized an atomic layer deposition (ALD) technique to generate an inorganic, silicon dioxide (SiO2) coating layer on chemically cross-linked PVA hydrogel fibers. To evaluate the stability of SiO2-coated PVA hydrogel fibers mimicking the in vivo environment, we conducted accelerated stability tests. SiO2-coated PVA hydrogel fibers showed improved stability over a one-week incubation period under a harsh environment, preventing swelling and preserving their mechanical and optical properties compared to uncoated fibers. These SiO2-coated PVA hydrogel fibers demonstrated nanoscale polymeric crystalline domains (6.5 ± 0.1 nm), an elastic modulus of 73.7 ± 31.7 MPa, a maximum elongation of 113.6 ± 24.2%, and minimal light transmission loss (1.9 ± 0.2 dB cm-1). Lastly, we applied these SiO2-coated PVA hydrogel fibers in vivo to optically activate the motor cortex of transgenic Thy1::ChR2 mice during locomotor behavioral tests. This mouse cohort was genetically modified to express the light-sensitive ion channel, channelrhodopsin-2 (ChR2), and implanted with hydrogel fibers to deliver light to the motor cortex area (M2). Light stimulation via hydrogel fibers resulted in optogenetically modulated mouse locomotor behaviors, including increased contralateral rotation, mobility speeds, and travel distances.
Collapse
Affiliation(s)
- Sizhe Huang
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA.
| | | | - Iyanah Mehta
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA.
| | - Omri Yosfan
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA.
| | - Eunji Hong
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA.
| | - Anyang Wang
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Nianqiang Wu
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Qianbin Wang
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA.
| | - Siyuan Rao
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA.
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
22
|
Tomitaka A, Vashist A, Kolishetti N, Nair M. Machine learning assisted-nanomedicine using magnetic nanoparticles for central nervous system diseases. NANOSCALE ADVANCES 2023; 5:4354-4367. [PMID: 37638161 PMCID: PMC10448356 DOI: 10.1039/d3na00180f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023]
Abstract
Magnetic nanoparticles possess unique properties distinct from other types of nanoparticles developed for biomedical applications. Their unique magnetic properties and multifunctionalities are especially beneficial for central nervous system (CNS) disease therapy and diagnostics, as well as targeted and personalized applications using image-guided therapy and theranostics. This review discusses the recent development of magnetic nanoparticles for CNS applications, including Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, and drug addiction. Machine learning (ML) methods are increasingly applied towards the processing, optimization and development of nanomaterials. By using data-driven approach, ML has the potential to bridge the gap between basic research and clinical research. We review ML approaches used within the various stages of nanomedicine development, from nanoparticle synthesis and characterization to performance prediction and disease diagnosis.
Collapse
Affiliation(s)
- Asahi Tomitaka
- Department of Computer and Information Sciences, College of Natural and Applied Science, University of Houston-Victoria Texas 77901 USA
| | - Arti Vashist
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
- Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
- Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
- Institute of NeuroImmune Pharmacology, Centre for Personalized Nanomedicine, Herbert Wertheim College of Medicine, Florida International University Miami Florida 33199 USA
| |
Collapse
|
23
|
Jung YJ, Kim H, Cheong HK, Lim YB. Magnetic control of self-assembly and disassembly in organic materials. Nat Commun 2023; 14:3081. [PMID: 37248227 DOI: 10.1038/s41467-023-38846-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 05/18/2023] [Indexed: 05/31/2023] Open
Abstract
Because organic molecules and materials are generally insensitive or weakly sensitive to magnetic fields, a certain means to enhance their magnetic responsiveness needs to be exploited. Here we show a strategy to amplify the magnetic responsiveness of self-assembled peptide nanostructures by synergistically combining the concepts of perfect α-helix and rod-coil supramolecular building blocks. Firstly, we develop a monomeric, nonpolar, and perfect α-helix (MNP-helix). Then, we employ the MNP-helix as the rod block of rod-coil amphiphiles (rod-coils) because rod-coils are well-suited for fabricating responsive assemblies. We show that the self-assembly processes of the designed rod-coils and disassembly of rod-coil/DNA complexes can be controlled in a magnetically responsive manner using the relatively weak magnetic field provided by the ordinary neodymium magnet [0.07 ~ 0.25 Tesla (T)]. These results demonstrate that magnetically responsive organic assemblies usable under practical conditions can be realized by using rod-coil supramolecular building blocks containing constructively organized diamagnetic moieties.
Collapse
Affiliation(s)
- You-Jin Jung
- Department of Materials Science & Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Hyoseok Kim
- Department of Materials Science & Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea
| | - Hae-Kap Cheong
- Division of Magnetic Resonance, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Yong-Beom Lim
- Department of Materials Science & Engineering, Yonsei University, 50 Yonsei-ro, Seoul, 03722, Republic of Korea.
| |
Collapse
|
24
|
Benayas E, Espinosa A, Portolés MT, Vila-del Sol V, Morales MP, Serrano MC. Cellular and Molecular Processes Are Differently Influenced in Primary Neural Cells by Slight Changes in the Physicochemical Properties of Multicore Magnetic Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17726-17741. [PMID: 36976318 PMCID: PMC10103129 DOI: 10.1021/acsami.3c02729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/21/2023] [Indexed: 06/18/2023]
Abstract
Herein, we use two exemplary superparamagnetic iron oxide multicore nanoparticles (SPIONs) to illustrate the significant influence of slightly different physicochemical properties on the cellular and molecular processes that define SPION interplay with primary neural cells. Particularly, we have designed two different SPION structures, NFA (i.e., a denser multicore structure accompanied by a slightly less negative surface charge and a higher magnetic response) and NFD (i.e., a larger surface area and more negatively charged), and identified specific biological responses dependent on SPION type, concentration, exposure time, and magnetic actuation. Interestingly, NFA SPIONs display a higher cell uptake, likely driven by their less negative surface and smaller protein corona, more significantly impacting cell viability and complexity. The tight contact of both SPIONs with neural cell membranes results in the significant augmentation of phosphatidylcholine, phosphatidylserine, and sphingomyelin and the reduction of free fatty acids and triacylglycerides for both SPIONs. Nonetheless, NFD induces greater effects on lipids, especially under magnetic actuation, likely indicating a preferential membranal location and/or a tighter interaction with membrane lipids than NFA, in agreement with their lower cell uptake. From a functional perspective, these lipid changes correlate with an increase in plasma membrane fluidity, again larger for more negatively charged nanoparticles (NFD). Finally, the mRNA expression of iron-related genes such as Ireb-2 and Fth-1 remains unaltered, while TfR-1 is only detected in SPION-treated cells. Taken together, these results demonstrate the substantial impact that minor physicochemical differences of nanomaterials may exert in the specific targeting of cellular and molecular processes. A denser multicore structure generated by autoclave-based production is accompanied by a slight difference in surface charge and magnetic properties that become decisive for the biological impact of these SPIONs. Their capacity to markedly modify the lipidic cell content makes them attractive as lipid-targetable nanomedicines.
Collapse
Affiliation(s)
- Esther Benayas
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Ana Espinosa
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - M. Teresa Portolés
- Departamento
de Bioquímica y Biología Molecular, Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico
San Carlos (IdISSC), Madrid 28040, Spain
- CIBER
de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (IDSCIII), Madrid 28040, Spain
| | - Virginia Vila-del Sol
- Hospital
Nacional de Parapléjicos, Servicio
de Salud de Castilla-La Mancha (SESCAM), Finca de la Peraleda s/n, Toledo 45071, Spain
| | - M. Puerto Morales
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - María C. Serrano
- , Instituto de
Ciencia de Materiales de Madrid, Consejo Superior de
Investigaciones Científicas, calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| |
Collapse
|
25
|
Rodrigues AF, Rebelo C, Reis T, Simões S, Bernardino L, Peça J, Ferreira L. Engineering optical tools for remotely controlled brain stimulation and regeneration. Biomater Sci 2023; 11:3034-3050. [PMID: 36947145 DOI: 10.1039/d2bm02059a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2023]
Abstract
Neurological disorders are one of the world's leading medical and societal challenges due to the lack of efficacy of the first line treatment. Although pharmacological and non-pharmacological interventions have been employed with the aim of regulating neuronal activity and survival, they have failed to avoid symptom relapse and disease progression in the vast majority of patients. In the last 5 years, advanced drug delivery systems delivering bioactive molecules and neuromodulation strategies have been developed to promote tissue regeneration and remodel neuronal circuitry. However, both approaches still have limited spatial and temporal precision over the desired target regions. While external stimuli such as electromagnetic fields and ultrasound have been employed in the clinic for non-invasive neuromodulation, they do not have the capability of offering single-cell spatial resolution as light stimulation. Herein, we review the latest progress in this area of study and discuss the prospects of using light-responsive nanomaterials to achieve on-demand delivery of drugs and neuromodulation, with the aim of achieving brain stimulation and regeneration.
Collapse
Affiliation(s)
- Artur Filipe Rodrigues
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Catarina Rebelo
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Tiago Reis
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Susana Simões
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Liliana Bernardino
- Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, 6201-506 Covilhã, Portugal
| | - João Peça
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| | - Lino Ferreira
- Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-517 Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, 3000-354 Coimbra, Portugal
- Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central, University of Coimbra, 3000-354 Coimbra, Portugal.
| |
Collapse
|
26
|
Xiong H, Alberto KA, Youn J, Taura J, Morstein J, Li X, Wang Y, Trauner D, Slesinger PA, Nielsen SO, Qin Z. Optical control of neuronal activities with photoswitchable nanovesicles. NANO RESEARCH 2023; 16:1033-1041. [PMID: 37063114 PMCID: PMC10103898 DOI: 10.1007/s12274-022-4853-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 06/19/2023]
Abstract
Precise modulation of neuronal activity by neuroactive molecules is essential for understanding brain circuits and behavior. However, tools for highly controllable molecular release are lacking. Here, we developed a photoswitchable nanovesicle with azobenzene-containing phosphatidylcholine (azo-PC), coined 'azosome', for neuromodulation. Irradiation with 365 nm light triggers the trans-to-cis isomerization of azo-PC, resulting in a disordered lipid bilayer with decreased thickness and cargo release. Irradiation with 455 nm light induces reverse isomerization and switches the release off. Real-time fluorescence imaging shows controllable and repeatable cargo release within seconds (< 3 s). Importantly, we demonstrate that SKF-81297, a dopamine D1-receptor agonist, can be repeatedly released from the azosome to activate cultures of primary striatal neurons. Azosome shows promise for precise optical control over the molecular release and can be a valuable tool for molecular neuroscience studies.
Collapse
Affiliation(s)
- Hejian Xiong
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Kevin A. Alberto
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jonghae Youn
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jaume Taura
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Johannes Morstein
- Department of Chemistry, New York University, New York, NY 10012, USA
| | - Xiuying Li
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Yang Wang
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Dirk Trauner
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul A. Slesinger
- Department of Chemistry, New York University, New York, NY 10012, USA
| | - Steven O. Nielsen
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Surgery, University of Texas at Southwestern Medical Center, Dallas, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
27
|
Xiong H, Alberto KA, Youn J, Taura J, Morstein J, Li X, Wang Y, Trauner D, Slesinger PA, Nielsen SO, Qin Z. Optical control of neuronal activities with photoswitchable nanovesicles. NANO RESEARCH 2023; 16:1033-1041. [PMID: 37063114 DOI: 10.1007/s12274-022-4976-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 05/25/2023]
Abstract
Precise modulation of neuronal activity by neuroactive molecules is essential for understanding brain circuits and behavior. However, tools for highly controllable molecular release are lacking. Here, we developed a photoswitchable nanovesicle with azobenzene-containing phosphatidylcholine (azo-PC), coined 'azosome', for neuromodulation. Irradiation with 365 nm light triggers the trans-to-cis isomerization of azo-PC, resulting in a disordered lipid bilayer with decreased thickness and cargo release. Irradiation with 455 nm light induces reverse isomerization and switches the release off. Real-time fluorescence imaging shows controllable and repeatable cargo release within seconds (< 3 s). Importantly, we demonstrate that SKF-81297, a dopamine D1-receptor agonist, can be repeatedly released from the azosome to activate cultures of primary striatal neurons. Azosome shows promise for precise optical control over the molecular release and can be a valuable tool for molecular neuroscience studies.
Collapse
Affiliation(s)
- Hejian Xiong
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Kevin A Alberto
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jonghae Youn
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Jaume Taura
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Johannes Morstein
- Department of Chemistry, New York University, New York, NY 10012, USA
| | - Xiuying Li
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Yang Wang
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Dirk Trauner
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul A Slesinger
- Department of Chemistry, New York University, New York, NY 10012, USA
| | - Steven O Nielsen
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Surgery, University of Texas at Southwestern Medical Center, Dallas, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
28
|
Central Nervous System Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
29
|
Gong X, Jiang Z, Zeng W, Hu C, Luo X, Lei W, Yuan C. Alternating Magnetic Field Induced Magnetic Heating in Ferromagnetic Cobalt Single-Atom Catalysts for Efficient Oxygen Evolution Reaction. NANO LETTERS 2022; 22:9411-9417. [PMID: 36410739 DOI: 10.1021/acs.nanolett.2c03359] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alternating magnetic field (AMF) is a promising methodology for further improving magnetic single-atom catalyst (SAC) activity toward oxygen evolution reaction (OER). Herein, the anchoring of Co single atoms on MoS2 support (Co@MoS2), leading to the appearance of in-plane room-temperature ferromagnetic properties, is favorable for the parallel spin arrangement of oxygen atoms when a magnetic field is applied. Moreover, field-assisted electrocatalytic experiments confirmed that the spin direction of Co@MoS2 is changing with the applied magnetic field. On this basis, under AMF, the active sites in ferromagnetic Co@MoS2 were heated by exploiting the magnetic heating generated from spin polarization flip of these SACs to further expedite OER efficiency, with overpotential at 10 mA cm-2 reduced from 317 mV to 250 mV. This work introduces a feasible and efficient approach to enhance the OER performance of Co@MoS2 by AMF, shedding some light on the further development of magnetic SACs for energy conversion.
Collapse
Affiliation(s)
- Xunguo Gong
- Jiangxi Key Laboratory of Nanomaterials and Sensors, School of Physics, Communication and Electronics, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, Jiangxi, China
| | - Zhenzhen Jiang
- Jiangxi Key Laboratory of Nanomaterials and Sensors, School of Physics, Communication and Electronics, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, Jiangxi, China
| | - Wei Zeng
- Jiangxi Key Laboratory of Nanomaterials and Sensors, School of Physics, Communication and Electronics, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, Jiangxi, China
| | - Ce Hu
- Jiangxi Key Laboratory of Nanomaterials and Sensors, School of Physics, Communication and Electronics, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, Jiangxi, China
| | - Xingfang Luo
- Jiangxi Key Laboratory of Nanomaterials and Sensors, School of Physics, Communication and Electronics, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, Jiangxi, China
| | - Wen Lei
- Department of Electrical, Electronic and Computer Engineering, The University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia
| | - Cailei Yuan
- Jiangxi Key Laboratory of Nanomaterials and Sensors, School of Physics, Communication and Electronics, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, Jiangxi, China
| |
Collapse
|
30
|
Doxorubicin Loaded Thermosensitive Magneto-Liposomes Obtained by a Gel Hydration Technique: Characterization and In Vitro Magneto-Chemotherapeutic Effect Assessment. Pharmaceutics 2022; 14:pharmaceutics14112501. [PMID: 36432692 PMCID: PMC9697793 DOI: 10.3390/pharmaceutics14112501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
The combination of magnetic hyperthermia with chemotherapy is considered a promising strategy in cancer therapy due to the synergy between the high temperatures and the chemotherapeutic effects, which can be further developed for targeted and remote-controlled drug release. In this paper we report a simple, rapid, and reproducible method for the preparation of thermosensitive magnetoliposomes (TsMLs) loaded with doxorubicin (DOX), consisting of a lipidic gel formation from a previously obtained water-in-oil microemulsion with fine aqueous droplets containing magnetic nanoparticles (MNPs) dispersed in an organic solution of thermosensitive lipids (transition temperature of ~43 °C), followed by the gel hydration with an aqueous solution of DOX. The obtained thermosensitive magnetoliposomes (TsMLs) were around 300 nm in diameter and exhibited 40% DOX incorporation efficiency. The most suitable MNPs to incorporate into the liposomal aqueous lumen were Zn ferrites, with a very low coercive field at 300 K (7 kA/m) close to the superparamagnetic regime, exhibiting a maximum absorption rate (SAR) of 1130 W/gFe when dispersed in water and 635 W/gFe when confined inside TsMLs. No toxicity of Zn ferrite MNPs or of TsMLs was noticed against the A459 cancer cell line after 48 h incubation over the tested concentration range. The passive release of DOX from the TsMLs after 48h incubation induced a toxicity starting with a dosage level of 62.5 ug/cm2. Below this threshold, the subsequent exposure to an alternating magnetic field (20-30 kA/m, 355 kHz) for 30 min drastically reduced the viability of the A459 cells due to the release of incorporated DOX. Our results strongly suggest that TsMLs represent a viable strategy for anticancer therapies using the magnetic field-controlled release of DOX.
Collapse
|
31
|
Romero G, Park J, Koehler F, Pralle A, Anikeeva P. Modulating cell signalling in vivo with magnetic nanotransducers. NATURE REVIEWS. METHODS PRIMERS 2022; 2:92. [PMID: 38111858 PMCID: PMC10727510 DOI: 10.1038/s43586-022-00170-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 12/20/2023]
Abstract
Weak magnetic fields offer nearly lossless transmission of signals within biological tissue. Magnetic nanomaterials are capable of transducing magnetic fields into a range of biologically relevant signals in vitro and in vivo. These nanotransducers have recently enabled magnetic control of cellular processes, from neuronal firing and gene expression to programmed apoptosis. Effective implementation of magnetically controlled cellular signalling relies on careful tailoring of magnetic nanotransducers and magnetic fields to the responses of the intended molecular targets. This primer discusses the versatility of magnetic modulation modalities and offers practical guidelines for selection of appropriate materials and field parameters, with a particular focus on applications in neuroscience. With recent developments in magnetic instrumentation and nanoparticle chemistries, including those that are commercially available, magnetic approaches promise to empower research aimed at connecting molecular and cellular signalling to physiology and behaviour in untethered moving subjects.
Collapse
Affiliation(s)
- Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Jimin Park
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florian Koehler
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnd Pralle
- Department of Physics, University at Buffalo, the State University of New York, Buffalo, NY, USA
| | - Polina Anikeeva
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
32
|
Xia X, Yin Z, Yang Y, Li S, Wang L, Cai X, Xu Y, Ma C, Qiu Y, Chen Z, Tan W. In Situ Upregulating Heat Shock Protein 70 via Gastric Nano-Heaters for the Interference of Helicobacter pylori Infection. ACS NANO 2022; 16:14043-14054. [PMID: 35993384 DOI: 10.1021/acsnano.2c03911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Taking inspiration from the mechanism of Helicobacter pylori infection can lead to innovative antibacterial ways to fight antibiotics resistance. Herein, a gastric nano-heater iron-cobalt alloy shielded with graphitic shells (FeCo@G) is developed to interfere with H. pylori infection under an alternating magnetic field. FeCo@G shows a high and stable specific loss power (SLP = 534.1 W g-1) in the acidic environment and provides efficient magnetothermal stimulation in the stomach. Such stimulation upregulates the cytoprotective heat shock protein 70 (HSP70) in gastric epithelial cells, which antagonizes the infection of H. pylori. This finding is further supported by the transcriptomic analysis verifying the upregulation of HSP70 in the stomach. Moreover, the nano-heater shows a high inhibition rate of H. pylori in vivo with good biocompatibility; 95% of FeCo@G is excreted from the mouse's gastrointestinal tract within 12 h. In summary, FeCo@G allows magnetothermal therapy to be used in harsh gastric environments, providing an approach for the therapy against H. pylori.
Collapse
Affiliation(s)
- Xin Xia
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Zhiwei Yin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Yanxia Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Shengkai Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Xinqi Cai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Yiting Xu
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan 411201, People's Republic of China
| | - Chao Ma
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Ye Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, People's Republic of China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|
33
|
Ko MJ, Hong H, Choi H, Kang H, Kim D. Multifunctional Magnetic Nanoparticles for Dynamic Imaging and Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Min Jun Ko
- Department of Radiology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
| | - Hyunsik Hong
- Department of Materials Science and Engineering Korea University Seoul 02841 Republic of Korea
| | - Hyunjun Choi
- Department of Radiology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
- Department of Bioengineering University of Illinois at Chicago Chicago IL 60607 USA
| | - Heemin Kang
- Department of Materials Science and Engineering Korea University Seoul 02841 Republic of Korea
- College of Medicine Korea University Seoul 02841 Republic of Korea
| | - Dong‐Hyun Kim
- Department of Radiology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
- Department of Bioengineering University of Illinois at Chicago Chicago IL 60607 USA
- Department of Biomedical Engineering McCormick School of Engineering Northwestern University Evanston IL 60208 USA
- Robert H. Lurie Comprehensive Cancer Center Northwestern University Chicago Illinois 60611 USA
| |
Collapse
|
34
|
Guntnur RT, Muzzio N, Gomez A, Macias S, Galindo A, Ponce A, Romero G. On-Demand Chemomagnetic Modulation of Striatal Neurons Facilitated by Hybrid Magnetic Nanoparticles. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2204732. [PMID: 36339020 PMCID: PMC9635318 DOI: 10.1002/adfm.202204732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Indexed: 06/15/2023]
Abstract
Minimally invasive manipulation of cell signaling is critical in basic neuroscience research and in developing therapies for neurological disorders. Here, we describe a wireless chemomagnetic neuromodulation platform for the on-demand control of primary striatal neurons that relies on nanoscale heating events. Iron oxide magnetic nanoparticles (MNPs) are functionally coated with thermoresponsive poly (oligo (ethylene glycol) methyl ether methacrylate) (POEGMA) brushes loaded with dopamine. Dopamine loaded MNPs-POEGMA are co-cultured with primary striatal neurons. When alternating magnetinec fields (AMF) are applied, MNPs undergo hysteresis power loss and dissipate heat. The local heat produced by MNPs initiates a thermodynamic phase transition on POEGMA brushes resulting in polymer collapse and dopamine release. AMF-triggered dopamine release enhances the response of dopamine ion channels expressed on the cell membranes enhancing the activity of ~50% of striatal neurons subjected to the treatment. Chemomagnetic actuation on dopamine receptors is confirmed by blocking D1 and D2 receptors. The reversible thermodynamic phase transition of POEGMA brushes allow the on-demand release of dopamine in multiple microdoses. AMF-triggered dopamine release from MNPs-POEGMA causes no cell cytotoxicity nor promotes cell ROS production. This research represents a fundamental step forward for the chemomagnetic control of neural activity using hybrid magnetic nanomaterials with tailored physical properties.
Collapse
Affiliation(s)
- Rohini Thevi Guntnur
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| | - Nicolas Muzzio
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| | - Amanda Gomez
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| | - Sean Macias
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| | - Arturo Galindo
- Department of Physics and Astronomy, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| | - Arturo Ponce
- Department of Physics and Astronomy, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio; San Antonio, TX 78249, USA
| |
Collapse
|
35
|
Progress, Opportunities, and Challenges of Magneto-Plasmonic Nanoparticles under Remote Magnetic and Light Stimulation for Brain-Tissue and Cellular Regeneration. NANOMATERIALS 2022; 12:nano12132242. [PMID: 35808077 PMCID: PMC9268050 DOI: 10.3390/nano12132242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 01/25/2023]
Abstract
Finding curable therapies for neurodegenerative disease (ND) is still a worldwide medical and clinical challenge. Recently, investigations have been made into the development of novel therapeutic techniques, and examples include the remote stimulation of nanocarriers to deliver neuroprotective drugs, genes, growth factors, and antibodies using a magnetic field and/or low-power lights. Among these potential nanocarriers, magneto-plasmonic nanoparticles possess obvious advantages, such as the functional restoration of ND models, due to their unique nanostructure and physiochemical properties. In this review, we provide an overview of the latest advances in magneto-plasmonic nanoparticles, and the associated therapeutic approaches to repair and restore brain tissues. We have reviewed their potential as smart nanocarriers, including their unique responsivity under remote magnetic and light stimulation for the controlled and sustained drug delivery for reversing neurodegenerations, as well as the utilization of brain organoids in studying the interaction between NPs and neuronal tissue. This review aims to provide a comprehensive summary of the current progress, opportunities, and challenges of using these smart nanocarriers for programmable therapeutics to treat ND, and predict the mechanism and future directions.
Collapse
|
36
|
Maeng LY, Rosenfeld D, Simandl GJ, Koehler F, Senko AW, Moon J, Varnavides G, Murillo MF, Reimer AE, Wald A, Anikeeva P, Widge AS. Probing Neuro-Endocrine Interactions Through Remote Magnetothermal Adrenal Stimulation. Front Neurosci 2022; 16:901108. [PMID: 35837128 PMCID: PMC9274974 DOI: 10.3389/fnins.2022.901108] [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: 03/21/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
Exposure to stressful or traumatic stimuli may alter hypothalamic-pituitary-adrenal (HPA) axis and sympathoadrenal-medullary (SAM) reactivity. This altered reactivity may be a component or cause of mental illnesses. Dissecting these mechanisms requires tools to reliably probe HPA and SAM function, particularly the adrenal component, with temporal precision. We previously demonstrated magnetic nanoparticle (MNP) technology to remotely trigger adrenal hormone release by activating thermally sensitive ion channels. Here, we applied adrenal magnetothermal stimulation to probe stress-induced HPA axis and SAM changes. MNP and control nanoparticles were injected into the adrenal glands of outbred rats subjected to a tone-shock conditioning/extinction/recall paradigm. We measured MNP-triggered adrenal release before and after conditioning through physiologic (heart rate) and serum (epinephrine, corticosterone) markers. Aversive conditioning altered adrenal function, reducing corticosterone and blunting heart rate increases post-conditioning. MNP-based organ stimulation provides a novel approach to probing the function of SAM, HPA, and other neuro-endocrine axes and could help elucidate changes across stress and disease models.
Collapse
Affiliation(s)
- Lisa Y. Maeng
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Dekel Rosenfeld
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Gregory J. Simandl
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Florian Koehler
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Alexander W. Senko
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Junsang Moon
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Georgios Varnavides
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Maria F. Murillo
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Adriano E. Reimer
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Aaron Wald
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Polina Anikeeva
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
- *Correspondence: Polina Anikeeva,
| | - Alik S. Widge
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, United States
- Alik S. Widge,
| |
Collapse
|
37
|
Swanson JL, Chin PS, Romero JM, Srivastava S, Ortiz-Guzman J, Hunt PJ, Arenkiel BR. Advancements in the Quest to Map, Monitor, and Manipulate Neural Circuitry. Front Neural Circuits 2022; 16:886302. [PMID: 35719420 PMCID: PMC9204427 DOI: 10.3389/fncir.2022.886302] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
Neural circuits and the cells that comprise them represent the functional units of the brain. Circuits relay and process sensory information, maintain homeostasis, drive behaviors, and facilitate cognitive functions such as learning and memory. Creating a functionally-precise map of the mammalian brain requires anatomically tracing neural circuits, monitoring their activity patterns, and manipulating their activity to infer function. Advancements in cell-type-specific genetic tools allow interrogation of neural circuits with increased precision. This review provides a broad overview of recombination-based and activity-driven genetic targeting approaches, contemporary viral tracing strategies, electrophysiological recording methods, newly developed calcium, and voltage indicators, and neurotransmitter/neuropeptide biosensors currently being used to investigate circuit architecture and function. Finally, it discusses methods for acute or chronic manipulation of neural activity, including genetically-targeted cellular ablation, optogenetics, chemogenetics, and over-expression of ion channels. With this ever-evolving genetic toolbox, scientists are continuing to probe neural circuits with increasing resolution, elucidating the structure and function of the incredibly complex mammalian brain.
Collapse
Affiliation(s)
- Jessica L. Swanson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Pey-Shyuan Chin
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Juan M. Romero
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Snigdha Srivastava
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Joshua Ortiz-Guzman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Patrick J. Hunt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Benjamin R. Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
38
|
Improvement of synaptic plasticity by nanoparticles and the related mechanisms: Applications and prospects. J Control Release 2022; 347:143-163. [PMID: 35513209 DOI: 10.1016/j.jconrel.2022.04.049] [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: 01/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/20/2022]
Abstract
Synaptic plasticity is an important basis of learning and memory and participates in brain network remodelling after different types of brain injury (such as that caused by neurodegenerative diseases, cerebral ischaemic injury, posttraumatic stress disorder (PTSD), and psychiatric disorders). Therefore, improving synaptic plasticity is particularly important for the treatment of nervous system-related diseases. With the rapid development of nanotechnology, increasing evidence has shown that nanoparticles (NPs) can cross the blood-brain barrier (BBB) in different ways, directly or indirectly act on nerve cells, regulate synaptic plasticity, and ultimately improve nerve function. Therefore, to better elucidate the effect of NPs on synaptic plasticity, we review evidence showing that NPs can improve synaptic plasticity by regulating different influencing factors, such as neurotransmitters, receptors, presynaptic membrane proteins and postsynaptic membrane proteins, and further discuss the possible mechanism by which NPs improve synaptic plasticity. We conclude that NPs can improve synaptic plasticity and restore the function of damaged nerves by inhibiting neuroinflammation and oxidative stress, inducing autophagy, and regulating ion channels on the cell membrane. By reviewing the mechanism by which NPs regulate synaptic plasticity and the applications of NPs for the treatment of neurological diseases, we also propose directions for future research in this field and provide an important reference for follow-up research.
Collapse
|
39
|
Zou L, Xu K, Tian H, Fang Y. Remote neural regulation mediated by nanomaterials. NANOTECHNOLOGY 2022; 33:272002. [PMID: 35442216 DOI: 10.1088/1361-6528/ac62b1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Neural regulation techniques play an essential role in the functional dissection of neural circuits and also the treatment of neurological diseases. Recently, a series of nanomaterials, including upconversion nanoparticles (UCNPs), magnetic nanoparticles (MNPs), and silicon nanomaterials (SNMs) that are responsive to remote optical or magnetic stimulation, have been applied as transducers to facilitate localized control of neural activities. In this review, we summarize the latest advances in nanomaterial-mediated neural regulation, especially in a remote and minimally invasive manner. We first give an overview of existing neural stimulation techniques, including electrical stimulation, transcranial magnetic stimulation, chemogenetics, and optogenetics, with an emphasis on their current limitations. Then we focus on recent developments in nanomaterial-mediated neural regulation, including UCNP-mediated fiberless optogenetics, MNP-mediated magnetic neural regulation, and SNM-mediated non-genetic neural regulation. Finally, we discuss the possibilities and challenges for nanomaterial-mediated neural regulation.
Collapse
Affiliation(s)
- Liang Zou
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ke Xu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huihui Tian
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
| | - Ying Fang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
40
|
Claes M, De Groef L, Moons L. The DREADDful Hurdles and Opportunities of the Chronic Chemogenetic Toolbox. Cells 2022; 11:1110. [PMID: 35406674 PMCID: PMC8998042 DOI: 10.3390/cells11071110] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/10/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022] Open
Abstract
The chronic character of chemogenetics has been put forward as one of the assets of the technique, particularly in comparison to optogenetics. Yet, the vast majority of chemogenetic studies have focused on acute applications, while repeated, long-term neuromodulation has only been booming in the past few years. Unfortunately, together with the rising number of studies, various hurdles have also been uncovered, especially in relation to its chronic application. It becomes increasingly clear that chronic neuromodulation warrants caution and that the effects of acute neuromodulation cannot be extrapolated towards chronic experiments. Deciphering the underlying cellular and molecular causes of these discrepancies could truly unlock the chronic chemogenetic toolbox and possibly even pave the way for chemogenetics towards clinical application. Indeed, we are only scratching the surface of what is possible with chemogenetic research. For example, most investigations are concentrated on behavioral read-outs, whereas dissecting the underlying molecular signature after (chronic) neuromodulation could reveal novel insights in terms of basic neuroscience and deregulated neural circuits. In this review, we highlight the hurdles associated with the use of chemogenetic experiments, as well as the unexplored research questions for which chemogenetics offers the ideal research platform, with a particular focus on its long-term application.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
| | - Lies De Groef
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
- Laboratory of Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
41
|
Ning S, Jorfi M, Patel SR, Kim DY, Tanzi RE. Neurotechnological Approaches to the Diagnosis and Treatment of Alzheimer’s Disease. Front Neurosci 2022; 16:854992. [PMID: 35401082 PMCID: PMC8989850 DOI: 10.3389/fnins.2022.854992] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in the elderly, clinically defined by progressive cognitive decline and pathologically, by brain atrophy, neuroinflammation, and accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles. Neurotechnological approaches, including optogenetics and deep brain stimulation, have exploded as new tools for not only the study of the brain but also for application in the treatment of neurological diseases. Here, we review the current state of AD therapeutics and recent advancements in both invasive and non-invasive neurotechnologies that can be used to ameliorate AD pathology, including neurostimulation via optogenetics, photobiomodulation, electrical stimulation, ultrasound stimulation, and magnetic neurostimulation, as well as nanotechnologies employing nanovectors, magnetic nanoparticles, and quantum dots. We also discuss the current challenges in developing these neurotechnological tools and the prospects for implementing them in the treatment of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Shen Ning
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Graduate Program for Neuroscience, Boston University School of Medicine, Boston, MA, United States
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Mehdi Jorfi,
| | - Shaun R. Patel
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Rudolph E. Tanzi,
| |
Collapse
|
42
|
Saha R, Wu K, Bloom RP, Liang S, Tonini D, Wang JP. A review on magnetic and spintronic neurostimulation: challenges and prospects. NANOTECHNOLOGY 2022; 33:182004. [PMID: 35013010 DOI: 10.1088/1361-6528/ac49be] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
In the treatment of neurodegenerative, sensory and cardiovascular diseases, electrical probes and arrays have shown quite a promising success rate. However, despite the outstanding clinical outcomes, their operation is significantly hindered by non-selective control of electric fields. A promising alternative is micromagnetic stimulation (μMS) due to the high permeability of magnetic field through biological tissues. The induced electric field from the time-varying magnetic field generated by magnetic neurostimulators is used to remotely stimulate neighboring neurons. Due to the spatial asymmetry of the induced electric field, high spatial selectivity of neurostimulation has been realized. Herein, some popular choices of magnetic neurostimulators such as microcoils (μcoils) and spintronic nanodevices are reviewed. The neurostimulator features such as power consumption and resolution (aiming at cellular level) are discussed. In addition, the chronic stability and biocompatibility of these implantable neurostimulator are commented in favor of further translation to clinical settings. Furthermore, magnetic nanoparticles (MNPs), as another invaluable neurostimulation material, has emerged in recent years. Thus, in this review we have also included MNPs as a remote neurostimulation solution that overcomes physical limitations of invasive implants. Overall, this review provides peers with the recent development of ultra-low power, cellular-level, spatially selective magnetic neurostimulators of dimensions within micro- to nano-range for treating chronic neurological disorders. At the end of this review, some potential applications of next generation neuro-devices have also been discussed.
Collapse
Affiliation(s)
- Renata Saha
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Kai Wu
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Robert P Bloom
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Shuang Liang
- Department of Chemical Engineering and Material Science, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Denis Tonini
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Jian-Ping Wang
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, United States of America
| |
Collapse
|
43
|
Abstract
Current technological advances in neural probing and modulation have enabled an extraordinary glimpse into the intricacies of the nervous system. Particularly, nanomaterials are proving to be an incredibly versatile platform for neurological applications owing to their biocompatibility, tunability, highly specific targeting and sensing, and long-term chemical stability. Among the most desirable nanomaterials for neuroengineering, freestanding nanomaterials are minimally invasive and remotely controlled. This review outlines the most recent developments of freestanding nanomaterials that operate on the neuronal interface. First, the different nanomaterials and their mechanisms for modulating neurons are explored to provide a basis for how freestanding nanomaterials operate. Then, the three main applications of subcellular neuronal engineering-modulating neuronal behavior, exploring fundamental neuronal mechanism, and recording neuronal signal-are highlighted with specific examples of current advancements. Finally, we conclude with our perspective on future nanomaterial designs and applications.
Collapse
Affiliation(s)
- Elaine Liang
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Jiuyun Shi
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
- The James Franck Institute, University of Chicago, Chicago, IL 60637, USA
- The Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Bozhi Tian
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
- The James Franck Institute, University of Chicago, Chicago, IL 60637, USA
- The Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
44
|
Fan H. Central Nervous System Nanotechnology. Nanomedicine (Lond) 2022. [DOI: 10.1007/978-981-13-9374-7_29-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
45
|
Lin YC, Fang TY, Kao HY, Tseng WC. Nanoassembly of UCST polypeptide for NIR-modulated drug release. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
46
|
Yang X, McGlynn E, Das R, Paşca SP, Cui B, Heidari H. Nanotechnology Enables Novel Modalities for Neuromodulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103208. [PMID: 34668249 PMCID: PMC8712412 DOI: 10.1002/adma.202103208] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/27/2021] [Indexed: 05/18/2023]
Abstract
Neuromodulation is of great importance both as a fundamental neuroscience research tool for analyzing and understanding the brain function, and as a therapeutic avenue for treating brain disorders. Here, an overview of conceptual and technical progress in developing neuromodulation strategies is provided, and it is suggested that recent advances in nanotechnology are enabling novel neuromodulation modalities with less invasiveness, improved biointerfaces, deeper penetration, and higher spatiotemporal precision. The use of nanotechnology and the employment of versatile nanomaterials and nanoscale devices with tailored physical properties have led to considerable research progress. To conclude, an outlook discussing current challenges and future directions for next-generation neuromodulation modalities is presented.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCA94305USA
- Stanford Brain OrganogenesisWu Tsai Neurosciences InstituteStanford UniversityStanfordCA94305USA
- Wu Tsai Neurosciences InstituteStanford UniversityStanfordCA94305USA
- Department of ChemistryStanford UniversityStanfordCA94305USA
| | - Eve McGlynn
- Microelectronics Lab (meLAB)James Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUK
| | - Rupam Das
- Microelectronics Lab (meLAB)James Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUK
| | - Sergiu P. Paşca
- Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCA94305USA
- Stanford Brain OrganogenesisWu Tsai Neurosciences InstituteStanford UniversityStanfordCA94305USA
| | - Bianxiao Cui
- Wu Tsai Neurosciences InstituteStanford UniversityStanfordCA94305USA
- Department of ChemistryStanford UniversityStanfordCA94305USA
| | - Hadi Heidari
- Microelectronics Lab (meLAB)James Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUK
| |
Collapse
|
47
|
Kolesov DV, Sokolinskaya EL, Lukyanov KA, Bogdanov AM. Molecular Tools for Targeted Control of Nerve Cell Electrical Activity. Part II. Acta Naturae 2021; 13:17-32. [PMID: 35127143 PMCID: PMC8807539 DOI: 10.32607/actanaturae.11415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 05/14/2021] [Indexed: 01/01/2023] Open
Abstract
In modern life sciences, the issue of a specific, exogenously directed manipulation of a cell's biochemistry is a highly topical one. In the case of electrically excitable cells, the aim of the manipulation is to control the cells' electrical activity, with the result being either excitation with subsequent generation of an action potential or inhibition and suppression of the excitatory currents. The techniques of electrical activity stimulation are of particular significance in tackling the most challenging basic problem: figuring out how the nervous system of higher multicellular organisms functions. At this juncture, when neuroscience is gradually abandoning the reductionist approach in favor of the direct investigation of complex neuronal systems, minimally invasive methods for brain tissue stimulation are becoming the basic element in the toolbox of those involved in the field. In this review, we describe three approaches that are based on the delivery of exogenous, genetically encoded molecules sensitive to external stimuli into the nervous tissue. These approaches include optogenetics (overviewed in Part I), as well as chemogenetics and thermogenetics (described here, in Part II), which is significantly different not only in the nature of the stimuli and structure of the appropriate effector proteins, but also in the details of experimental applications. The latter circumstance is an indication that these are rather complementary than competing techniques.
Collapse
Affiliation(s)
- D. V. Kolesov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - E. L. Sokolinskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - K. A. Lukyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - A. M. Bogdanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| |
Collapse
|
48
|
Wunderlich H, Kozielski KL. Next generation material interfaces for neural engineering. Curr Opin Biotechnol 2021; 72:29-38. [PMID: 34601203 DOI: 10.1016/j.copbio.2021.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/06/2021] [Accepted: 09/07/2021] [Indexed: 11/28/2022]
Abstract
Neural implant technology is rapidly progressing, and gaining broad interest in research fields such as electrical engineering, materials science, neurobiology, and data science. As the potential applications of neural devices have increased, new technologies to make neural intervention longer-lasting and less invasive have brought attention to neural interface engineering. This review will focus on recent developments in materials for neural implants, highlighting new technologies in the fields of soft electrodes, mechanical and chemical engineering of interface coatings, and remotely powered devices. In this context, novel implantation strategies, manufacturing methods, and combinatorial device functions will also be discussed.
Collapse
Affiliation(s)
- Hannah Wunderlich
- Department of Bioengineering and Biosystems, Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kristen L Kozielski
- Department of Electrical and Computer Engineering, Technical University of Munich, Munich, Germany.
| |
Collapse
|
49
|
Jiménez GL, Thevi Guntnur R, Guiliani J, Romero G. Enhancing magnetic hyperthermia in ferrite nanoparticles through shape anisotropy and surface hybridization. AIChE J 2021. [DOI: 10.1002/aic.17437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Gloria L. Jiménez
- Department of Biomedical Engineering and Chemical Engineering University of Texas at San Antonio. One UTSA Circle San Antonio Texas USA
| | - Rohini Thevi Guntnur
- Department of Biomedical Engineering and Chemical Engineering University of Texas at San Antonio. One UTSA Circle San Antonio Texas USA
| | - Jason Guiliani
- Department of Physics and Astronomy University of Texas at San Antonio. One UTSA Circle San Antonio Texas USA
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering University of Texas at San Antonio. One UTSA Circle San Antonio Texas USA
| |
Collapse
|
50
|
Hu X, Li F, Xia F, Wang Q, Lin P, Wei M, Gong L, Low LE, Lee JY, Ling D. Dynamic nanoassembly-based drug delivery system (DNDDS): Learning from nature. Adv Drug Deliv Rev 2021; 175:113830. [PMID: 34139254 DOI: 10.1016/j.addr.2021.113830] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/19/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Dynamic nanoassembly-based drug delivery system (DNDDS) has evolved from being a mere curiosity to emerging as a promising strategy for high-performance diagnosis and/or therapy of various diseases. However, dynamic nano-bio interaction between DNDDS and biological systems remains poorly understood, which can be critical for precise spatiotemporal and functional control of DNDDS in vivo. To deepen the understanding for fine control over DNDDS, we aim to explore natural systems as the root of inspiration for researchers from various fields. This review highlights ingenious designs, nano-bio interactions, and controllable functionalities of state-of-the-art DNDDS under endogenous or exogenous stimuli, by learning from nature at the molecular, subcellular, and cellular levels. Furthermore, the assembly strategies and response mechanisms of tailor-made DNDDS based on the characteristics of various diseased microenvironments are intensively discussed. Finally, the current challenges and future perspectives of DNDDS are briefly commented.
Collapse
|