1
|
Eldar D, Albert S, Tatyana A, Galina S, Albert R, Yana M. Optogenetic approaches for neural tissue regeneration: A review of basic optogenetic principles and target cells for therapy. Neural Regen Res 2026; 21:521-533. [PMID: 39995064 DOI: 10.4103/nrr.nrr-d-24-00685] [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: 06/21/2024] [Accepted: 10/17/2024] [Indexed: 02/26/2025] Open
Abstract
Optogenetics has revolutionized the field of neuroscience by enabling precise control of neural activity through light-sensitive proteins known as opsins. This review article discusses the fundamental principles of optogenetics, including the activation of both excitatory and inhibitory opsins, as well as the development of optogenetic models that utilize recombinant viral vectors. A considerable portion of the article addresses the limitations of optogenetic tools and explores strategies to overcome these challenges. These strategies include the use of adeno-associated viruses, cell-specific promoters, modified opsins, and methodologies such as bioluminescent optogenetics. The application of viral recombinant vectors, particularly adeno-associated viruses, is emerging as a promising avenue for clinical use in delivering opsins to target cells. This trend indicates the potential for creating tools that offer greater flexibility and accuracy in opsin delivery. The adaptations of these viral vectors provide advantages in optogenetic studies by allowing for the restricted expression of opsins through cell-specific promoters and various viral serotypes. The article also examines different cellular targets for optogenetics, including neurons, astrocytes, microglia, and Schwann cells. Utilizing specific promoters for opsin expression in these cells is essential for achieving precise and efficient stimulation. Research has demonstrated that optogenetic stimulation of both neurons and glial cells-particularly the distinct phenotypes of microglia, astrocytes, and Schwann cells-can have therapeutic effects in neurological diseases. Glial cells are increasingly recognized as important targets for the treatment of these disorders. Furthermore, the article emphasizes the emerging field of bioluminescent optogenetics, which combines optogenetic principles with bioluminescent proteins to visualize and manipulate neural activity in real time. By integrating molecular genetics techniques with bioluminescence, researchers have developed methods to monitor neuronal activity efficiently and less invasively, enhancing our understanding of central nervous system function and the mechanisms of plasticity in neurological disorders beyond traditional neurobiological methods. Evidence has shown that optogenetic modulation can enhance motor axon regeneration, achieve complete sensory reinnervation, and accelerate the recovery of neuromuscular function. This approach also induces complex patterns of coordinated motor neuron activity and promotes neural reorganization. Optogenetic approaches hold immense potential for therapeutic interventions in the central nervous system. They enable precise control of neural circuits and may offer new treatments for neurological disorders, particularly spinal cord injuries, peripheral nerve injuries, and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Davletshin Eldar
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Sufianov Albert
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Research and Educational Institute of Neurosurgery, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Ageeva Tatyana
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Sufianova Galina
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Rizvanov Albert
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Mukhamedshina Yana
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| |
Collapse
|
2
|
Kale MB, Wankhede NL, Bishoyi AK, Ballal S, Kalia R, Arya R, Kumar S, Khalid M, Gulati M, Umare M, Taksande BG, Upaganlawar AB, Umekar MJ, Kopalli SR, Fareed M, Koppula S. Emerging biophysical techniques for probing synaptic transmission in neurodegenerative disorders. Neuroscience 2025; 565:63-79. [PMID: 39608699 DOI: 10.1016/j.neuroscience.2024.11.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Plethora of research has shed light on the critical role of synaptic dysfunction in various neurodegenerative disorders (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Synapses, the fundamental units for neural communication in the brain, are highly vulnerable to pathological conditions and are central to the progression of neurological diseases. The presynaptic terminal, a key component of synapses responsible for neurotransmitter release and synaptic communication, undergoes structural and functional alterations in these disorders. Understanding synaptic transmission abnormalities is crucial for unravelling the pathophysiological mechanisms underlying neurodegeneration. In the quest to probe synaptic transmission in NDDs, emerging biophysical techniques play a pivotal role. These advanced methods offer insights into the structural and functional changes occurring at nerve terminals in conditions like AD, PD, HD & ALS. By investigating synaptic plasticity and alterations in neurotransmitter release dynamics, researchers can uncover valuable information about disease progression and potential therapeutic targets. The review articles highlighted provide a comprehensive overview of how synaptic vulnerability and pathology are shared mechanisms across a spectrum of neurological disorders. In major neurodegenerative diseases, synaptic dysfunction is a common thread linking these conditions. The intricate molecular machinery involved in neurotransmitter release, synaptic vesicle dynamics, and presynaptic protein regulation are key areas of focus for understanding synaptic alterations in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Mohit Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box- 71666, Riyadh 11597, Saudi Arabia
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
3
|
Livingston CA, Weng CY, Chancellor JR. Retinitis Pigmentosa and Therapeutic Candidates. Int Ophthalmol Clin 2025; 65:17-21. [PMID: 39710900 DOI: 10.1097/iio.0000000000000542] [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: 12/24/2024]
Abstract
Retinitis pigmentosa (RP) is a class of inherited retinal dystrophies (IRDs) that involves the degeneration of retinal photoreceptor cells and results in progressive vision loss. It was identified and named in 1857. For over 100 years, treatment of RP was generally limited to modifications in diet, management of cystoid macular edema, and supportive care for low vision. Over the last several decades, advances in technology and our understanding of the human genome have led to a host of new therapeutic candidates for the treatment of RP. This includes gene and cell therapy, optogenetics, neuroprotective agents, and electronic retinal implants. In this article, we summarize both the traditional and novel therapeutic modalities for the treatment of retinitis pigmentosa.
Collapse
|
4
|
Chen D, Zhao Z, Shi J, Li S, Xu X, Wu Z, Tang Y, Liu N, Zhou W, Ni C, Ma B, Wang J, Zhang J, Huang L, You Z, Zhang P, Tang Z. Harnessing the sensing and stimulation function of deep brain-machine interfaces: a new dawn for overcoming substance use disorders. Transl Psychiatry 2024; 14:440. [PMID: 39419976 PMCID: PMC11487193 DOI: 10.1038/s41398-024-03156-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Substance use disorders (SUDs) imposes profound physical, psychological, and socioeconomic burdens on individuals, families, communities, and society as a whole, but the available treatment options remain limited. Deep brain-machine interfaces (DBMIs) provide an innovative approach by facilitating efficient interactions between external devices and deep brain structures, thereby enabling the meticulous monitoring and precise modulation of neural activity in these regions. This pioneering paradigm holds significant promise for revolutionizing the treatment landscape of addictive disorders. In this review, we carefully examine the potential of closed-loop DBMIs for addressing SUDs, with a specific emphasis on three fundamental aspects: addictive behaviors-related biomarkers, neuromodulation techniques, and control policies. Although direct empirical evidence is still somewhat limited, rapid advancements in cutting-edge technologies such as electrophysiological and neurochemical recordings, deep brain stimulation, optogenetics, microfluidics, and control theory offer fertile ground for exploring the transformative potential of closed-loop DBMIs for ameliorating symptoms and enhancing the overall well-being of individuals struggling with SUDs.
Collapse
Affiliation(s)
- Danyang Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhixian Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jian Shi
- School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shengjie Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xinran Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuojin Wu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenhong Zhou
- Wuhan Global Sensor Technology Co., Ltd, Wuhan, Hubei, China
| | - Changmao Ni
- Wuhan Neuracom Technology Development Co., Ltd, Wuhan, Hubei, China
| | - Bo Ma
- Microsystems Technology Center, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junya Wang
- Microsystems Technology Center, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Zhang
- School of Electrical Engineering and Automation, Wuhan University, Wuhan, Hubei, China
| | - Li Huang
- Wuhan Neuracom Technology Development Co., Ltd, Wuhan, Hubei, China
| | - Zheng You
- Microsystems Technology Center, School of Mechanical Science and Engineering, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
5
|
Li Y, Yang B, Wang Y, Huang Z, Wang J, Pu X, Wen J, Ao Q, Xiao K, Wu J, Yin G. Postoperatively Noninvasive Optogenetic Stimulation via Upconversion Nanoparticles Enhancing Sciatic Nerve Repair. NANO LETTERS 2024; 24:5403-5412. [PMID: 38669639 DOI: 10.1021/acs.nanolett.3c04619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The efficacy of electrical stimulation facilitating peripheral nerve regeneration is evidenced extensively, while the associated secondary damage resulting from repeated electrode invasion and indiscriminate stimulation is inevitable. Here, we present an optogenetics strategy that utilizes upconversion nanoparticles (UCNPs) to convert deeply penetrating near-infrared excitation into blue emission, which activates an adeno-associated virus-encoding ChR2 photoresponsive ion channel on cell membranes. The induced Ca2+ flux, similar to the ion flux in the electrical stimulation approach, efficiently regulates viability and proliferation, secretion of nerve growth factor, and neural function of RSC96 cells. Furthermore, deep near-infrared excitation is harnessed to stimulate autologous Schwann cells in situ via a UCNP-composited scaffold, which enhances nerve sprouting and myelination, consequently promoting functional recovery, electrophysiological restoration, and reinnervation of damaged nerves. This developed postoperatively noninvasive optogenetics strategy presents a novel, minimally traumatic, and enduring therapeutic stimulus to effectively promote peripheral nerve repair.
Collapse
Affiliation(s)
- Ya Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, China
| | - Bing Yang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Precision Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610093, China
| | - Yulin Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Jirui Wen
- Department of Otolaryngology Head and Neck Surgery/Deep Underground Space Medical Center West China Hospital, Sichuan University, No. 37 Guoxuexiang, Chengdu 610041, China
| | - Qiang Ao
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, China
| | - Kai Xiao
- Precision Medicine Research Center of West China Hospital, Sichuan University, Chengdu 610093, China
| | - Jiang Wu
- Department of Otolaryngology Head and Neck Surgery/Deep Underground Space Medical Center West China Hospital, Sichuan University, No. 37 Guoxuexiang, Chengdu 610041, China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
6
|
Faltus T, Freise J, Fluck C, Zillmann H. Ethics and regulation of neuronal optogenetics in the European Union. Pflugers Arch 2023; 475:1505-1517. [PMID: 37996706 PMCID: PMC10730653 DOI: 10.1007/s00424-023-02888-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Neuronal optogenetics is a technique to control the activity of neurons with light. This is achieved by artificial expression of light-sensitive ion channels in the target cells. By optogenetic methods, cells that are naturally light-insensitive can be made photosensitive and addressable by illumination and precisely controllable in time and space. So far, optogenetics has primarily been a basic research tool to better understand the brain. However, initial studies are already investigating the possibility of using optogenetics in humans for future therapeutic approaches for neuronal based diseases such as Parkinson's disease, epilepsy, or to promote stroke recovery. In addition, optogenetic methods have already been successfully applied to a human in an experimental setting. Neuronal optogenetics also raises ethical and legal issues, e.g., in relation to, animal experiments, and its application in humans. Additional ethical and legal questions may arise when optogenetic methods are investigated on cerebral organoids. Thus, for the successful translation of optogenetics from basic research to medical practice, the ethical and legal questions of this technology must also be answered, because open ethical and legal questions can hamper the translation. The paper provides an overview of the ethical and legal issues raised by neuronal optogenetics. In addition, considering the technical prerequisites for translation, the paper shows consistent approaches to address these open questions. The paper also aims to support the interdisciplinary dialogue between scientists and physicians on the one hand, and ethicists and lawyers on the other, to enable an interdisciplinary coordinated realization of neuronal optogenetics.
Collapse
Affiliation(s)
- Timo Faltus
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Johannes Freise
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Carsten Fluck
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hans Zillmann
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany.
| |
Collapse
|
7
|
Li J, Wu C, Zeng M, Zhang Y, Wei D, Sun J, Fan H. Functional material-mediated wireless physical stimulation for neuro-modulation and regeneration. J Mater Chem B 2023; 11:9056-9083. [PMID: 37649427 DOI: 10.1039/d3tb01354e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Nerve injuries and neurological diseases remain intractable clinical challenges. Despite the advantages of stem cell therapy in treating neurological disorders, uncontrollable cell fates and loss of cell function in vivo are still challenging. Recently, increasing attention has been given to the roles of external physical signals, such as electricity and ultrasound, in regulating stem cell fate as well as activating or inhibiting neuronal activity, which provides new insights for the treatment of neurological disorders. However, direct physical stimulations in vivo are short in accuracy and safety. Functional materials that can absorb energy from a specific physical field exerted in a wireless way and then release another localized physical signal hold great advantages in mediating noninvasive or minimally invasive accurate indirect physical stimulations to promote the therapeutic effect on neurological disorders. In this review, the mechanism by which various physical signals regulate stem cell fate and neuronal activity is summarized. Based on these concepts, the approaches of using functional materials to mediate indirect wireless physical stimulation for neuro-modulation and regeneration are systematically reviewed. We expect that this review will contribute to developing wireless platforms for neural stimulation as an assistance for the treatment of neurological diseases and injuries.
Collapse
Affiliation(s)
- Jialu Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, Sichuan, China
| | - Mingze Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
8
|
Xu S, Momin M, Ahmed S, Hossain A, Veeramuthu L, Pandiyan A, Kuo CC, Zhou T. Illuminating the Brain: Advances and Perspectives in Optoelectronics for Neural Activity Monitoring and Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303267. [PMID: 37726261 DOI: 10.1002/adma.202303267] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/30/2023] [Indexed: 09/21/2023]
Abstract
Optogenetic modulation of brain neural activity that combines optical and electrical modes in a unitary neural system has recently gained robust momentum. Controlling illumination spatial coverage, designing light-activated modulators, and developing wireless light delivery and data transmission are crucial for maximizing the use of optical neuromodulation. To this end, biocompatible electrodes with enhanced optoelectrical performance, device integration for multiplexed addressing, wireless transmission, and multimodal operation in soft systems have been developed. This review provides an outlook for uniformly illuminating large brain areas while spatiotemporally imaging the neural responses upon optoelectrical stimulation with little artifacts. Representative concepts and important breakthroughs, such as head-mounted illumination, multiple implanted optical fibers, and micro-light-delivery devices, are discussed. Examples of techniques that incorporate electrophysiological monitoring and optoelectrical stimulation are presented. Challenges and perspectives are posed for further research efforts toward high-density optoelectrical neural interface modulation, with the potential for nonpharmacological neurological disease treatments and wireless optoelectrical stimulation.
Collapse
Affiliation(s)
- Shumao Xu
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Marzia Momin
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Salahuddin Ahmed
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Arafat Hossain
- Department of Electrical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Loganathan Veeramuthu
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Archana Pandiyan
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Chi-Ching Kuo
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Tao Zhou
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| |
Collapse
|
9
|
Chen F, Qian J, Cao Z, Li A, Cui J, Shi L, Xie J. Chemogenetic and optogenetic stimulation of zona incerta GABAergic neurons ameliorates motor impairment in Parkinson's disease. iScience 2023; 26:107149. [PMID: 37416450 PMCID: PMC10319825 DOI: 10.1016/j.isci.2023.107149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/26/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons in the substantia nigra and leads to progressive motor dysfunction. While studies have focused on the basal ganglia network, recent evidence suggests neuronal systems outside the basal ganglia are also related to PD pathogenesis. The zona incerta (ZI) is a predominantly inhibitory subthalamic region for global behavioral modulation. This study investigates the role of GABAergic neurons in the ZI in a mouse model of 6-hydroxydopamine (6-OHDA)-induced PD. First, we found a decrease in GABA-positive neurons in the ZI, and then the mice used chemogenetic/optogenetic stimulation to activate or inhibit GABAergic neurons. The motor performance of PD mice was significantly improved by chemogenetic/optogenetic activation of GABAergic neurons, and repeated chemogenetic activation of ZI GABAergic neurons increased the dopamine content in the striatum. Our work identifies the role of ZI GABAergic neurons in regulating motor behaviors in 6-OHDA-lesioned PD model mice.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Junliang Qian
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Zhongkai Cao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Ang Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Juntao Cui
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Limin Shi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, School of Basic Medicine, Qingdao University, Qingdao, China
- Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Goyal D, Kumar H. In Vivo and 3D Imaging Technique(s) for Spatiotemporal Mapping of Pathological Events in Experimental Model(s) of Spinal Cord Injury. ACS Chem Neurosci 2023; 14:809-819. [PMID: 36787542 DOI: 10.1021/acschemneuro.2c00643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Endothelial damage, astrogliosis, microgliosis, and neuronal degeneration are the most common events after spinal cord injury (SCI). Studies highlighted that studying the spatiotemporal profile of these events might provide a deeper understanding of the pathophysiology of SCI. For imaging of these events, available conventional techniques such as 2-dimensional histology and immunohistochemistry (IHC) are well established and frequently used to visualize and detect the altered expression of the protein of interest involved in these events. However, the technique requires the physical sectioning of the tissue, and results are also open to misinterpretation. Currently, researchers are focusing more attention toward the advanced tools for imaging the spinal cord's various physiological and pathological parameters. The tools include two-photon imaging, light sheet fluorescence microscopy, in vivo imaging system with fluorescent probes, and in vivo chemical and fluorescent protein-expressing viral-tracers. These techniques outperform the limitations associated with conventional techniques in various aspects, such as optical sectioning of tissue, 3D reconstructed imaging, and imaging of particular planes of interest. In addition to this, these techniques are minimally invasive and less time-consuming. In this review, we will discuss the various advanced imaging methodologies that will evolve in the future to explore the fundamental mechanisms after SCI.
Collapse
Affiliation(s)
- Divya Goyal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat India, 382355
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat India, 382355
| |
Collapse
|
11
|
Liu W, Liu S, Li P, Yao K. Retinitis Pigmentosa: Progress in Molecular Pathology and Biotherapeutical Strategies. Int J Mol Sci 2022; 23:ijms23094883. [PMID: 35563274 PMCID: PMC9101511 DOI: 10.3390/ijms23094883] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/13/2022] Open
Abstract
Retinitis pigmentosa (RP) is genetically heterogeneous retinopathy caused by photoreceptor cell death and retinal pigment epithelial atrophy that eventually results in blindness in bilateral eyes. Various photoreceptor cell death types and pathological phenotypic changes that have been disclosed in RP demand in-depth research of its pathogenic mechanism that may account for inter-patient heterogeneous responses to mainstream drug treatment. As the primary method for studying the genetic characteristics of RP, molecular biology has been widely used in disease diagnosis and clinical trials. Current technology iterations, such as gene therapy, stem cell therapy, and optogenetics, are advancing towards precise diagnosis and clinical applications. Specifically, technologies, such as effective delivery vectors, CRISPR/Cas9 technology, and iPSC-based cell transplantation, hasten the pace of personalized precision medicine in RP. The combination of conventional therapy and state-of-the-art medication is promising in revolutionizing RP treatment strategies. This article provides an overview of the latest research on the pathogenesis, diagnosis, and treatment of retinitis pigmentosa, aiming for a convenient reference of what has been achieved so far.
Collapse
|
12
|
Kim S, Nam Y, Kim HS, Jung H, Jeon SG, Hong SB, Moon M. Alteration of Neural Pathways and Its Implications in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10040845. [PMID: 35453595 PMCID: PMC9025507 DOI: 10.3390/biomedicines10040845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease accompanied by cognitive and behavioral symptoms. These AD-related manifestations result from the alteration of neural circuitry by aggregated forms of amyloid-β (Aβ) and hyperphosphorylated tau, which are neurotoxic. From a neuroscience perspective, identifying neural circuits that integrate various inputs and outputs to determine behaviors can provide insight into the principles of behavior. Therefore, it is crucial to understand the alterations in the neural circuits associated with AD-related behavioral and psychological symptoms. Interestingly, it is well known that the alteration of neural circuitry is prominent in the brains of patients with AD. Here, we selected specific regions in the AD brain that are associated with AD-related behavioral and psychological symptoms, and reviewed studies of healthy and altered efferent pathways to the target regions. Moreover, we propose that specific neural circuits that are altered in the AD brain can be potential targets for AD treatment. Furthermore, we provide therapeutic implications for targeting neuronal circuits through various therapeutic approaches and the appropriate timing of treatment for AD.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Hyeon soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
- Correspondence:
| |
Collapse
|
13
|
Mirzayi P, Shobeiri P, Kalantari A, Perry G, Rezaei N. Optogenetics: implications for Alzheimer's disease research and therapy. Mol Brain 2022; 15:20. [PMID: 35197102 PMCID: PMC8867657 DOI: 10.1186/s13041-022-00905-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD), a critical neurodegenerative condition, has a wide range of effects on brain activity. Synaptic plasticity and neuronal circuits are the most vulnerable in Alzheimer's disease, but the exact mechanism is unknown. Incorporating optogenetics into the study of AD has resulted in a significant leap in this field during the last decades, kicking off a revolution in our knowledge of the networks that underpin cognitive functions. In Alzheimer's disease, optogenetics can help to reduce and reverse neural circuit and memory impairments. Here we review how optogenetically driven methods have helped expand our knowledge of Alzheimer's disease, and how optogenetic interventions hint at a future translation into therapeutic possibilities for further utilization in clinical settings. In conclusion, neuroscience has witnessed one of its largest revolutions following the introduction of optogenetics into the field.
Collapse
Affiliation(s)
- Parsa Mirzayi
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Kalantari
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
14
|
Zheng N, Fitzpatrick V, Cheng R, Shi L, Kaplan DL, Yang C. Photoacoustic Carbon Nanotubes Embedded Silk Scaffolds for Neural Stimulation and Regeneration. ACS NANO 2022; 16:2292-2305. [PMID: 35098714 DOI: 10.1021/acsnano.1c08491] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Neural interfaces using biocompatible scaffolds provide crucial properties, such as cell adhesion, structural support, and mass transport, for the functional repair of nerve injuries and neurodegenerative diseases. Neural stimulation has also been found to be effective in promoting neural regeneration. This work provides a generalized strategy to integrate photoacoustic (PA) neural stimulation into hydrogel scaffolds using a nanocomposite hydrogel approach. Specifically, polyethylene glycol (PEG)-functionalized carbon nanotubes (CNT), highly efficient photoacoustic agents, are embedded into silk fibroin to form biocompatible and soft photoacoustic materials. We show that these photoacoustic functional scaffolds enable nongenetic activation of neurons with a spatial precision defined by the area of light illumination, promoting neuron regeneration. These CNT/silk scaffolds offered reliable and repeatable photoacoustic neural stimulation, and 94% of photoacoustic-stimulated neurons exhibit a fluorescence change larger than 10% in calcium imaging in the light-illuminated area. The on-demand photoacoustic stimulation increased neurite outgrowth by 1.74-fold in a rat dorsal root ganglion model, when compared to the unstimulated group. We also confirmed that promoted neurite outgrowth by photoacoustic stimulation is associated with an increased concentration of neurotrophic factor (BDNF). As a multifunctional neural scaffold, CNT/silk scaffolds demonstrated nongenetic PA neural stimulation functions and promoted neurite outgrowth, providing an additional method for nonpharmacological neural regeneration.
Collapse
Affiliation(s)
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | | | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | | |
Collapse
|
15
|
Affiliation(s)
- Harald Janovjak
- EMBL Australia, Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
| | | |
Collapse
|
16
|
Keshmiri Neghab H, Soheilifar MH, Grusch M, Ortega MM, Esmaeeli Djavid G, Saboury AA, Goliaei B. The state of the art of biomedical applications of optogenetics. Lasers Surg Med 2021; 54:202-216. [PMID: 34363230 DOI: 10.1002/lsm.23463] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Optogenetics has opened new insights into biomedical research with the ability to manipulate and control cellular activity using light in combination with genetically engineered photosensitive proteins. By stimulating with light, this method provides high spatiotemporal and high specificity resolution, which is in contrast to conventional pharmacological or electrical stimulation. Optogenetics was initially introduced to control neural activities but was gradually extended to other biomedical fields. STUDY DESIGN In this paper, firstly, we summarize the current optogenetic tools stimulated by different light sources, including lasers, light-emitting diodes, and laser diodes. Second, we outline the variety of biomedical applications of optogenetics not only for neuronal circuits but also for various kinds of cells and tissues from cardiomyocytes to ganglion cells. Furthermore, we highlight the potential of this technique for treating neurological disorders, cardiac arrhythmia, visual impairment, hearing loss, and urinary bladder diseases as well as clarify the mechanisms underlying cancer progression and control of stem cell differentiation. CONCLUSION We sought to summarize the various types of promising applications of optogenetics to treat a broad spectrum of disorders. It is conceivable to expect that optogenetics profits a growing number of patients suffering from a range of different diseases in the near future.
Collapse
Affiliation(s)
- Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | | | - Michael Grusch
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Manoela Marques Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, São Paulo, Brazil
| | - Gholamreza Esmaeeli Djavid
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Ali Akbar Saboury
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Bahram Goliaei
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
17
|
Fan CH, Wei KC, Chiu NH, Liao EC, Wang HC, Wu RY, Ho YJ, Chan HL, Wang TSA, Huang YZ, Hsieh TH, Lin CH, Lin YC, Yeh CK. Sonogenetic-Based Neuromodulation for the Amelioration of Parkinson's Disease. NANO LETTERS 2021; 21:5967-5976. [PMID: 34264082 DOI: 10.1021/acs.nanolett.1c00886] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sonogenetics is a promising strategy allowing the noninvasive and selective activation of targeted neurons in deep brain regions; nevertheless, its therapeutic outcome for neurodegeneration diseases that need long-term treatment remains to be verified. We previously enhanced the ultrasound (US) sensitivity of targeted cells by genetic modification with an engineered auditory-sensing protein, mPrestin (N7T, N308S). In this study, we expressed mPrestin in the dopaminergic neurons of the substantia nigra in Parkinson's disease (PD) mice and used 0.5 MHz US for repeated and localized brain stimulation. The mPrestin expression in dopaminergic neurons persisted for at least 56 days after a single shot of adeno-associated virus, suggesting that the period of expression was long enough for US treatment in mice. Compared to untreated mice, US stimulation ameliorated the dopaminergic neurodegeneration 10-fold and mitigated the PD symptoms of the mice 4-fold, suggesting that this sonogenetic strategy has the clinical potential to treat neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Kuo-Chen Wei
- New Taipei Municipal TuCheng Hospital, New Taipei City 236017, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan 33305, Taiwan
| | | | | | | | - Ruo-Yu Wu
- Department of Chemistry, National Taiwan University, Taipei 106319, Taiwan
| | | | | | | | | | | | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 106319, Taiwan
| | | | | |
Collapse
|
18
|
Audouard E, Rousselot L, Folcher M, Cartier N, Piguet F. Optimized Protocol for Subcutaneous Implantation of Encapsulated Cells Device and Evaluation of Biocompatibility. Front Bioeng Biotechnol 2021; 9:620967. [PMID: 34249877 PMCID: PMC8264370 DOI: 10.3389/fbioe.2021.620967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
Improving a drug delivery system is critical to treat central nervous system disorders. Here we studied an innovative approach based on implantation of a wireless-powered cell-based device in mice. This device, coupling biologic material and electronics, is the first of its kind. The advantage of this technology is its ability to control the secretion of a therapeutic molecule and to switch the classical permanent delivery to activation on demand. In diseases with relapsing-remitting phases such as multiple sclerosis, such activation could be selectively achieved in relapsing phases. However, the safety (tolerance to biomaterials and surgical procedure) of such a clinical device needs to be verified. Therefore, the development of tools to assess the biocompatibility of the system in animal models is an essential step. We present the development of this new therapeutic approach, the challenges we encountered during the different steps of its development (such as cell loading in the chamber, surgery protocol for subcutaneous implantation of the device) and the tools we used to evaluate cell viability and biocompatibility of the device.
Collapse
Affiliation(s)
- Emilie Audouard
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| | - Lisa Rousselot
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| | - Marc Folcher
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Basel, Switzerland.,Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland
| | - Nathalie Cartier
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| | - Françoise Piguet
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| |
Collapse
|
19
|
Oyigeya M. Reflex memory theory of acquired involuntary motor and sensory disorders. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2021. [DOI: 10.1186/s41983-021-00307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Explicit and implicit memories are conserved but flexible biological tools that nature uses to regulate the daily behaviors of human beings. An aberrant form of the implicit memory is presumed to exist and may be contributory to the pathophysiology of disorders such as tardive syndromes, phantom phenomena, flashback, posttraumatic stress disorders (PTSD), and related disorders. These disorders have posed significant clinical problems for both patients and physicians for centuries. All extant pathophysiological theories of these disorders have failed to provide basis for effective treatment.
Objective
The objective of this article is to propose an alternative pathophysiological theory that will hopefully lead to new treatment approaches.
Methods
The author sourced over 60 journal articles that treated topics on memory, and involuntary motor and sensory disorders, from open access journals using Google Scholar, and reviewed them and this helped in the formulation of this theory.
Results
From the reviews, the author thinks physical or chemical insult to the nervous system can cause defective circuit remodeling, leading to generation of a variant of implicit (automatic) memory, herein called “reflex memory” and this is encoded interoceptively to contribute to these phenomena states.
Conclusion
Acquired involuntary motor and sensory disorders are caused by defective circuit remodeling involving multiple neural mechanisms. Dysregulation of excitatory neurotransmitters, calcium overload, homeostatic failure, and neurotoxicity are implicated in the process. Sustained effects of these defective mechanisms are encoded interoceptively as abnormal memory in the neurons and the conscious manifestations are these disorders. Extant theories failed to recognize this possibility.
Collapse
|
20
|
Ingles-Prieto A, Furthmann N, Crossman SH, Tichy AM, Hoyer N, Petersen M, Zheden V, Biebl J, Reichhart E, Gyoergy A, Siekhaus DE, Soba P, Winklhofer KF, Janovjak H. Optogenetic delivery of trophic signals in a genetic model of Parkinson's disease. PLoS Genet 2021; 17:e1009479. [PMID: 33857132 PMCID: PMC8049241 DOI: 10.1371/journal.pgen.1009479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
Optogenetics has been harnessed to shed new mechanistic light on current and future therapeutic strategies. This has been to date achieved by the regulation of ion flow and electrical signals in neuronal cells and neural circuits that are known to be affected by disease. In contrast, the optogenetic delivery of trophic biochemical signals, which support cell survival and are implicated in degenerative disorders, has never been demonstrated in an animal model of disease. Here, we reengineered the human and Drosophila melanogaster REarranged during Transfection (hRET and dRET) receptors to be activated by light, creating one-component optogenetic tools termed Opto-hRET and Opto-dRET. Upon blue light stimulation, these receptors robustly induced the MAPK/ERK proliferative signaling pathway in cultured cells. In PINK1B9 flies that exhibit loss of PTEN-induced putative kinase 1 (PINK1), a kinase associated with familial Parkinson's disease (PD), light activation of Opto-dRET suppressed mitochondrial defects, tissue degeneration and behavioral deficits. In human cells with PINK1 loss-of-function, mitochondrial fragmentation was rescued using Opto-dRET via the PI3K/NF-кB pathway. Our results demonstrate that a light-activated receptor can ameliorate disease hallmarks in a genetic model of PD. The optogenetic delivery of trophic signals is cell type-specific and reversible and thus has the potential to inspire novel strategies towards a spatio-temporal regulation of tissue repair.
Collapse
Affiliation(s)
- Alvaro Ingles-Prieto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Nikolas Furthmann
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Samuel H. Crossman
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Nina Hoyer
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Petersen
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Zheden
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Julia Biebl
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Eva Reichhart
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Attila Gyoergy
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Daria E. Siekhaus
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Peter Soba
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstanze F. Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Harald Janovjak
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
- * E-mail:
| |
Collapse
|
21
|
Yi MH, Liu YU, Umpierre AD, Chen T, Ying Y, Zheng J, Dheer A, Bosco DB, Dong H, Wu LJ. Optogenetic activation of spinal microglia triggers chronic pain in mice. PLoS Biol 2021; 19:e3001154. [PMID: 33739978 PMCID: PMC8011727 DOI: 10.1371/journal.pbio.3001154] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/31/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022] Open
Abstract
Spinal microglia are highly responsive to peripheral nerve injury and are known to be a key player in pain. However, there has not been direct evidence showing that selective microglial activation in vivo is sufficient to induce chronic pain. Here, we used optogenetic approaches in microglia to address this question employing CX3CR1creER/+: R26LSL-ReaChR/+ transgenic mice, in which red-activated channelrhodopsin (ReaChR) is inducibly and specifically expressed in microglia. We found that activation of ReaChR by red light in spinal microglia evoked reliable inward currents and membrane depolarization. In vivo optogenetic activation of microglial ReaChR in the spinal cord triggered chronic pain hypersensitivity in both male and female mice. In addition, activation of microglial ReaChR up-regulated neuronal c-Fos expression and enhanced C-fiber responses. Mechanistically, ReaChR activation led to a reactive microglial phenotype with increased interleukin (IL)-1β production, which is likely mediated by inflammasome activation and calcium elevation. IL-1 receptor antagonist (IL-1ra) was able to reverse the pain hypersensitivity and neuronal hyperactivity induced by microglial ReaChR activation. Therefore, our work demonstrates that optogenetic activation of spinal microglia is sufficient to trigger chronic pain phenotypes by increasing neuronal activity via IL-1 signaling.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Yong U. Liu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Anthony D. Umpierre
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Yanlu Ying
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hailong Dong
- Department of Anesthesiology & Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, United States of America
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
22
|
Piguet F, de Saint Denis T, Audouard E, Beccaria K, André A, Wurtz G, Schatz R, Alves S, Sevin C, Zerah M, Cartier N. The Challenge of Gene Therapy for Neurological Diseases: Strategies and Tools to Achieve Efficient Delivery to the Central Nervous System. Hum Gene Ther 2021; 32:349-374. [PMID: 33167739 DOI: 10.1089/hum.2020.105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
For more than 10 years, gene therapy for neurological diseases has experienced intensive research growth and more recently therapeutic interventions for multiple indications. Beneficial results in several phase 1/2 clinical studies, together with improved vector technology have advanced gene therapy for the central nervous system (CNS) in a new era of development. Although most initial strategies have focused on orphan genetic diseases, such as lysosomal storage diseases, more complex and widespread conditions like Alzheimer's disease, Parkinson's disease, epilepsy, or chronic pain are increasingly targeted for gene therapy. Increasing numbers of applications and patients to be treated will require improvement and simplification of gene therapy protocols to make them accessible to the largest number of affected people. Although vectors and manufacturing are a major field of academic research and industrial development, there is a growing need to improve, standardize, and simplify delivery methods. Delivery is the major issue for CNS therapies in general, and particularly for gene therapy. The blood-brain barrier restricts the passage of vectors; strategies to bypass this obstacle are a central focus of research. In this study, we present the different ways that can be used to deliver gene therapy products to the CNS. We focus on results obtained in large animals that have allowed the transfer of protocols to human patients and have resulted in the generation of clinical data. We discuss the different routes of administration, their advantages, and their limitations. We describe techniques, equipment, and protocols and how they should be selected for safe delivery and improved efficiency for the next generation of gene therapy trials for CNS diseases.
Collapse
Affiliation(s)
- Françoise Piguet
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Timothée de Saint Denis
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Emilie Audouard
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Kevin Beccaria
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Arthur André
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Neurosurgery, Hôpitaux Universitaires La Pitié-Salpêtrière, Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Guillaume Wurtz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Raphael Schatz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Sandro Alves
- BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France
| | - Caroline Sevin
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France.,APHP, Department of Neurology, Hopital le Kremlin Bicetre, Paris, France
| | - Michel Zerah
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Nathalie Cartier
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
23
|
Ma K, Han XX, Yang XM, Zhou SL. Proteolysis targeting chimera technology: a novel strategy for treating diseases of the central nervous system. Neural Regen Res 2021; 16:1944-1949. [PMID: 33642364 PMCID: PMC8343312 DOI: 10.4103/1673-5374.308075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurological diseases such as stroke, Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease are among the intractable diseases for which appropriate drugs and treatments are lacking. Proteolysis targeting chimera (PROTAC) technology is a novel strategy to solve this problem. PROTAC technology uses the ubiquitin-protease system to eliminate mutated, denatured, and harmful proteins in cells. It can be reused, and utilizes the protein destruction mechanism of the cells, thus making up for the deficiencies of traditional protein degradation methods. It can effectively target and degrade proteins, including proteins that are difficult to identify and bind. Therefore, it has extremely important implications for drug development and the treatment of neurological diseases. At present, the targeted degradation of mutant BTK, mHTT, Tau, EGFR, and other proteins using PROTAC technology is gaining attention. It is expected that corresponding treatment of nervous system diseases can be achieved. This review first focuses on the recent developments in PROTAC technology in terms of protein degradation, drug production, and treatment of central nervous system diseases, and then discusses its limitations. This review will provide a brief overview of the recent application of PROTAC technology in the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Ke Ma
- College of Life Science, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Xiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Ming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Song-Lin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
24
|
Controlling the behaviour of Drosophila melanogaster via smartphone optogenetics. Sci Rep 2020; 10:17614. [PMID: 33077824 PMCID: PMC7572528 DOI: 10.1038/s41598-020-74448-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/24/2020] [Indexed: 01/05/2023] Open
Abstract
Invertebrates such as Drosophila melanogaster have proven to be a valuable model organism for studies of the nervous system. In order to control neuronal activity, optogenetics has evolved as a powerful technique enabling non-invasive stimulation using light. This requires light sources that can deliver patterns of light with high temporal and spatial precision. Currently employed light sources for stimulation of small invertebrates, however, are either limited in spatial resolution or require sophisticated and bulky equipment. In this work, we used smartphone displays for optogenetic control of Drosophila melanogaster. We developed an open-source smartphone app that allows time-dependent display of light patterns and used this to activate and inhibit different neuronal populations in both larvae and adult flies. Characteristic behavioural responses were observed depending on the displayed colour and brightness and in agreement with the activation spectra and light sensitivity of the used channelrhodopsins. By displaying patterns of light, we constrained larval movement and were able to guide larvae on the display. Our method serves as a low-cost high-resolution testbench for optogenetic experiments using small invertebrate species and is particularly appealing to application in neuroscience teaching labs.
Collapse
|
25
|
Du M, Huang L, Zheng J, Xi Y, Dai Y, Zhang W, Yan W, Tao G, Qiu J, So K, Ren C, Zhou S. Flexible Fiber Probe for Efficient Neural Stimulation and Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001410. [PMID: 32775173 PMCID: PMC7404151 DOI: 10.1002/advs.202001410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Indexed: 05/24/2023]
Abstract
Functional probes are a leading contender for the recognition and manipulation of nervous behavior and are characterized by substantial scientific and technological potential. Despite the recent development of functional neural probes, a flexible biocompatible probe unit that allows for long-term simultaneous stimulation and signaling is still an important task. Here, a category of flexible tiny multimaterial fiber probes (<0.3 g) is described in which the metal electrodes are regularly embedded inside a biocompatible polymer fiber with a double-clad optical waveguide by thermal drawing. Significantly, this arrangement enables great improvement in mechanical properties, achieves high optical transmission (>90%), and effectively minimizes the impedance (by up to one order of magnitude) of the probe. This ability allows to realize long-term (at least 10 weeks) simultaneous optical stimulation and neural recording at the single-cell level in behaving mice with signal-to-noise ratio (SNR = 30 dB) that is more than 6 times that of the benchmark probe such as an all-polymer fiber.
Collapse
Affiliation(s)
- Minghui Du
- State Key Laboratory of Luminescent Materials and DevicesSchool of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640China
- Guangdong Provincial Key Laboratory of Fibre Laser Materials and Applied TechniquesGuangdong Engineering Technology Research and Development Center of Special Optical Fibre Materials and DevicesGuangzhou510640China
| | - Lu Huang
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationMinistry of Education CNS Regeneration Collaborative Joint LaboratoryJinan UniversityGuangzhou510632China
- Department of Neurology and Stroke CenterThe First Affiliated Hospital of Jinan UniversityGuangzhou510632China
| | - Jiajun Zheng
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationMinistry of Education CNS Regeneration Collaborative Joint LaboratoryJinan UniversityGuangzhou510632China
| | - Yue Xi
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationMinistry of Education CNS Regeneration Collaborative Joint LaboratoryJinan UniversityGuangzhou510632China
| | - Yi Dai
- State Key Laboratory of Luminescent Materials and DevicesSchool of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640China
- Guangdong Provincial Key Laboratory of Fibre Laser Materials and Applied TechniquesGuangdong Engineering Technology Research and Development Center of Special Optical Fibre Materials and DevicesGuangzhou510640China
| | - Weida Zhang
- State Key Laboratory of Luminescent Materials and DevicesSchool of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640China
- Guangdong Provincial Key Laboratory of Fibre Laser Materials and Applied TechniquesGuangdong Engineering Technology Research and Development Center of Special Optical Fibre Materials and DevicesGuangzhou510640China
| | - Wei Yan
- Research Laboratory of ElectronicsMassachusetts Institute of Technology (MIT)CambridgeMA02139USA
| | - Guangming Tao
- School of Optical and Electronic InformationWuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhan430074China
| | - Jianrong Qiu
- College of Optical Science and EngineeringState Key Laboratory of Modern Optical InstrumentationZhejiang UniversityHangzhou310027China
| | - Kwok‐Fai So
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationMinistry of Education CNS Regeneration Collaborative Joint LaboratoryJinan UniversityGuangzhou510632China
| | - Chaoran Ren
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationMinistry of Education CNS Regeneration Collaborative Joint LaboratoryJinan UniversityGuangzhou510632China
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangzhou510530China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Center for Brain Science and Brain‐Inspired IntelligenceGuangdong‐Hong Kong‐Macao Greater Bay AreaGuangzhou510000China
| | - Shifeng Zhou
- State Key Laboratory of Luminescent Materials and DevicesSchool of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640China
- Guangdong Provincial Key Laboratory of Fibre Laser Materials and Applied TechniquesGuangdong Engineering Technology Research and Development Center of Special Optical Fibre Materials and DevicesGuangzhou510640China
| |
Collapse
|
26
|
Niyazi M, Zibaii MI, Chavoshinezhad S, Hamidabadi HG, Dargahi L, Bojnordi MN, Alizadeh R, Heravi M, Karimi H, Hosseini M, Sadeghi Malvajerdi E, Seyednazari M. Neurogenic differentiation of human dental pulp stem cells by optogenetics stimulation. J Chem Neuroanat 2020; 109:101821. [PMID: 32512152 DOI: 10.1016/j.jchemneu.2020.101821] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 05/09/2020] [Accepted: 06/01/2020] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Human dental pulp stem cells (hDPSCs), a promising source for autologous transplantation in regenerative medicine, have been shown to be able to differentiate into neural precursors. Optogenetics is considered as an advanced biological technique in neuroscience which is able to control the activity of genetically modified stem cells by light. The purpose of this study is to investigate the neurogenic differentiation of hDPSCs following optogenetic stimulation. METHODS The hDPSCs were isolated by mechanical enzymatic digestion from an impacted third molar and cultured in DMEM/F12. The cells were infected with lentiviruses carrying CaMKIIa-hChR2 (H134R). Opsin-expressing hDPSCs were plated at the density of 5 × 104 cells/well in 6-well plates and optical stimulation was conducted with blue light (470 nm) pulsing at 15 Hz, 90 % Duty Cycle and 10 mW power for 10 s every 90 minutes, 6 times a day for 5 days. Two control groups including non-opsin-expressing hDPSCs and opsin-expressing hDPSCs with no optical stimulation were also included in the study. A day after last light stimulation, the viability of cells was analyzed by the MTT assay and the morphological changes were examined by phase contrast microscopy. The expression of Nestin, Microtubule-Associated protein 2 (MAP2) and Doublecortin (DCX) were examined by immunocytochemistry. RESULTS Human DPSCs expressed the reporter gene, mCherry, 72 hours after lentiviral infection. The result of MTT assay revealed a significant more viability in optical stimulated opsin-expressing hDPSCs as compared with two control groups. Moreover, optical stimulation increased the expression of Nestin, Doublecortin and MAP2 along with morphological changes from spindle shape to neuron-like shape. CONCLUSION Optogenetics stimulation through depolarizing the hDPSCs can increase the cells viability and/or proliferation and also promote the differentiation toward neuron-like cells.
Collapse
Affiliation(s)
- Mahsa Niyazi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | | | - Sara Chavoshinezhad
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetic Research Center, Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Nazm Bojnordi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetic Research Center, Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Rafieh Alizadeh
- ENT and Head & Neck Research Center and Department, The Five Senses Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mansooreh Heravi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedieh Karimi
- Laser and Plasma Research Institute-Shahid Beheshti University, Tehran, Iran
| | - Mohammad Hosseini
- Laser and Plasma Research Institute-Shahid Beheshti University, Tehran, Iran
| | | | | |
Collapse
|
27
|
Bostancıklıoğlu M. An update on memory formation and retrieval: An engram-centric approach. Alzheimers Dement 2020; 16:926-937. [PMID: 32333509 DOI: 10.1002/alz.12071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/26/2019] [Accepted: 01/03/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVE We explore here that memory loss observed in the early stage of Alzheimer's disease (AD) is a disorder of memory retrieval, instead of a storage impairment. This engram-centric explanation aims to enlarge the conceptual frame of memory as an emergent behavior of the brain and to propose a new treatment strategy for memory retrieval in dementia-AD. BACKGROUND The conventional memory hypothesis suggests that memory is stored as multiple traces in hippocampal neurons but recent evidence indicates that there are specialized memory engrams responsible for the storage and the retrieval of different memory types. UPDATED MEMORY HYPOTHESIS There are specialized memory engram neurons for each memory type and when information will be stored as a memory arrives in the hippocampus through afferent neurons finds its neuron according to the excitability states of engram neurons. The excitability level in engram neurons seems like a code canalizing the interactions between engrams and information. Therefore, to enhance the excitability of memory engram neurons improves memory loss observed in AD. In addition, we suggest that the hippocampus creates an index for information stored in memory engram cells in specialized regions for different types of memory, instead of storing all information; and different anatomic locations of engram cells and their roles in memory retrieval point out that memory could be an emergent behavior of the brain, and the interaction between serotonin fluctuation and engram neurons could be neural underpinnings of terminal lucidity. MAJOR CHALLENGES FOR THE MODEL The major challenge for this engram-centric memory retrieval model is the translation from bench to patient, specifically the delivery of optogenetic tools in patients. Engram neurons can be specifically activated by optogenetic tools, but optogenetics is an invasive technique which requires optic fiber implantation into the brain. In addition, light can overheat the tissue and thus induce damage in tissue. Furthermore, light is a foreign object and its direct implantation into the brain may cause neuroinflammation, the main trigger of neurodegenerative diseases. Therefore, to test the engram hypothesis in human, new tools to allow specific engram activation should be discovered.
Collapse
|
28
|
Frequency-Specific Optogenetic Deep Brain Stimulation of Subthalamic Nucleus Improves Parkinsonian Motor Behaviors. J Neurosci 2020; 40:4323-4334. [PMID: 32312888 DOI: 10.1523/jneurosci.3071-19.2020] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 11/21/2022] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is an effective therapy for the motor symptoms of Parkinson's disease (PD). However, the neural elements mediating symptom relief are unclear. A previous study concluded that direct optogenetic activation of STN neurons was neither necessary nor sufficient for relief of parkinsonian symptoms. However, the kinetics of the channelrhodopsin-2 (ChR2) used for cell-specific activation are too slow to follow the high rates required for effective DBS, and thus the contribution of activation of STN neurons to the therapeutic effects of DBS remains unclear. We quantified the behavioral and neuronal effects of optogenetic STN DBS in female rats following unilateral 6-hydroxydopamine (6-OHDA) lesion using an ultrafast opsin (Chronos). Optogenetic STN DBS at 130 pulses per second (pps) reduced pathologic circling and ameliorated deficits in forelimb stepping similarly to electrical DBS, while optogenetic STN DBS with ChR2 did not produce behavioral effects. As with electrical DBS, optogenetic STN DBS exhibited a strong dependence on stimulation rate; high rates produced symptom relief while low rates were ineffective. High-rate optogenetic DBS generated both increases and decreases in firing rates of single neurons in STN, globus pallidus externa (GPe), and substantia nigra pars reticular (SNr), and disrupted β band oscillatory activity in STN and SNr. High-rate optogenetic STN DBS can indeed ameliorate parkinsonian motor symptoms through reduction of abnormal oscillatory activity in the STN-associated neural circuit, and these results highlight that the kinetic properties of opsins have a strong influence on the effects of optogenetic stimulation.SIGNIFICANCE STATEMENT Whether STN local cells contribute to the therapeutic effects of subthalamic nucleus (STN) deep brain stimulation (DBS) in Parkinson's disease (PD) remains unclear. We re-examined the role of STN local cells in mediating the symptom-relieving effects of STN DBS using cell type-specific optogenetic stimulation with a much faster opsin, Chronos. Direct optogenetic stimulation of STN neurons was effective in treating the symptoms of parkinsonism in the 6-hydroxydopamine (6-OHDA) lesion rat. These results highlight that the kinetic properties of opsins can have a strong influence on the effects of optogenetic activation/inhibition and must be considered when employing optogenetic to study high-rate neural stimulation.
Collapse
|
29
|
Wu CY, Fan CH, Chiu NH, Ho YJ, Lin YC, Yeh CK. Targeted delivery of engineered auditory sensing protein for ultrasound neuromodulation in the brain. Am J Cancer Res 2020; 10:3546-3561. [PMID: 32206107 PMCID: PMC7069068 DOI: 10.7150/thno.39786] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/08/2020] [Indexed: 12/26/2022] Open
Abstract
Sonogenetics is a promising approach for in vivo neuromodulation using ultrasound (US) to non-invasively stimulate cells in deep tissue. However, sonogenetics requires accurate transduction of US-responsive proteins into target cells. Here, we introduce a non-invasive and non-viral approach for intracerebral gene delivery. This approach utilizes temporary ultrasonic disruption of the blood-brain barrier (BBB) to transfect neurons at specific sites in the brain via DNA that encodes engineered US-responsive protein (murine Prestin (N7T, N308S))-loaded microbubbles (pPrestin-MBs). Prestin is a transmembrane protein that exists in the mammalian auditory system and functions as an electromechanical transducer. We further improved the US sensitivity of Prestin by introducing specific amino acid substitutions that frequently occur in sonar species into the mouse Prestin protein. We demonstrated this concept in mice using US with pPrestin-MBs to non-invasively modify and activate neurons within the brain for spatiotemporal neuromodulation. Method: MBs composed of cationic phospholipid and C3F8 loaded with mouse Prestin plasmid (pPrestin) via electrostatic interactions. The mean concentration and size of the pPrestin-MBs were (16.0 ± 0.2) × 109 MBs/mL and 1.1 ± 0.2 μm, respectively. SH-SY5Y neuron-like cells and C57BL mice were used in this study. We evaluated the gene transfection efficiency and BBB-opening region resulting from pPrestin-MBs with 1-MHz US (pressure = 0.1-0.5 MPa, cycle = 50-10000, pulse repetition frequency (PRF): 0.5-5 Hz, sonication time = 60 s) using green fluorescence protein (Venus) and Evans blue staining. Results: The maximum pPrestin expression with the highest cell viability occurred at a pressure of 0.5 MPa, cycle number of 5000, and PRF of 1 Hz. The cellular transfection rate with pPrestin-MBs and US was 20.2 ± 2.5%, which was 1.5-fold higher than that of commercial transfection agents (LT-1). In vivo data suggested that the most profound expression of pPrestin occurred at 2 days after performing pPrestin-MBs with US (0.5 MPa, 240 s sonication time). In addition, no server erythrocyte extravasations and apoptosis cells were observed at US-sonicated region. We further found that with 0.5-MHz US stimulation, cells with Prestin expression were 6-fold more likely to exhibit c-Fos staining than cells without Prestin expression. Conclusion: Successful activation of Prestin-expressing neurons suggests that this technology provides non-invasive and spatially precise selective modulation of one or multiple specific brain regions.
Collapse
|
30
|
CRISPR, Prime Editing, Optogenetics, and DREADDs: New Therapeutic Approaches Provided by Emerging Technologies in the Treatment of Spinal Cord Injury. Mol Neurobiol 2020; 57:2085-2100. [DOI: 10.1007/s12035-019-01861-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
|
31
|
Wang KW, Ye XL, Huang T, Yang XF, Zou LY. Optogenetics-induced activation of glutamate receptors improves memory function in mice with Alzheimer's disease. Neural Regen Res 2019; 14:2147-2155. [PMID: 31397354 PMCID: PMC6788230 DOI: 10.4103/1673-5374.262593] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/18/2019] [Indexed: 12/21/2022] Open
Abstract
Optogenetics is a combination of optics and genetics technology that can be used to activate or inhibit specific cells in tissues. It has been used to treat Parkinson's disease, epilepsy and neurological diseases, but rarely Alzheimer's disease. Adeno-associated virus carrying the CaMK promoter driving the optogenetic channelrhodopsin-2 (CHR2) gene (or without the CHR2 gene, as control) was injected into the bilateral dentate gyri, followed by repeated intrahippocampal injections of soluble low-molecular-weight amyloid-β1-42 peptide (Aβ1-42). Subsequently, the region was stimulated with a 473 nm laser (1-3 ms, 10 Hz, 5 minutes). The novel object recognition test was conducted to test memory function in mice. Immunohistochemical staining was performed to analyze the numbers of NeuN and synapsin Ia/b-positive cells in the hippocampus. Western blot assay was carried out to analyze the expression levels of glial fibrillary acidic protein, NeuN, synapsin Ia/b, metabotropic glutamate receptor-1a (mGluR-1a), mGluR-5, N-methyl-D-aspartate receptor subunit NR1, glutamate receptor 2, interleukin-1β, interleukin-6 and interleukin-10. Optogenetic stimulation improved working and short-term memory in mice with Alzheimer's disease. This neuroprotective effect was associated with increased expression of NR1, glutamate receptor 2 and mGluR-5 in the hippocampus, and decreased expression of glial fibrillary acidic protein and interleukin-6. Our results show that optogenetics can be used to regulate the neuronal-glial network to ameliorate memory functions in mice with Alzheimer's disease. The study was approved by the Animal Resources Committee of Jinan University, China (approval No. LL-KT-2011134) on February 28, 2011.
Collapse
Affiliation(s)
- Ke-Wei Wang
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| | - Xiao-Lin Ye
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| | - Ting Huang
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| | - Xi-Fei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong Province, China
| | - Liang-Yu Zou
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| |
Collapse
|
32
|
Shiri Z, Simorgh S, Naderi S, Baharvand H. Optogenetics in the Era of Cerebral Organoids. Trends Biotechnol 2019; 37:1282-1294. [PMID: 31227305 DOI: 10.1016/j.tibtech.2019.05.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
The human brain has been deemed the most complex organ and has captivated neuroscientists for decades. Most studies of this organ have relied on reductionist model systems. Although all model systems are essentially wrong, cerebral organoids so far represent the closest recapitulation of human brain development and disease both in terms of cell diversity and organization. The optogenetic technique can be used in this context to study the functional neuroanatomy of the brain, to examine the neural circuits, and to determine the etiology of neurological disorders. In this opinion article, we suggest ways in which optogenetics can be combined with cerebral organoids to allow unprecedented precision and accuracy in studying normal and aberrant neurodevelopmental processes and, as well, neurodegenerative diseases.
Collapse
Affiliation(s)
- Zahra Shiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Susan Simorgh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Somayeh Naderi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
33
|
Xie Z, Yang Q, Song D, Quan Z, Qing H. Optogenetic manipulation of astrocytes from synapses to neuronal networks: A potential therapeutic strategy for neurodegenerative diseases. Glia 2019; 68:215-226. [PMID: 31400164 DOI: 10.1002/glia.23693] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 02/06/2023]
Abstract
Astrocytes are the most widespread and heterogeneous glial cells in the central nervous system and key regulators for brain development. They are capable of receiving neurotransmitters produced by synaptic activities and regulating synaptic functions by releasing gliotransmitters as part of the tripartite synapse. In addition to communicating with neurons at synaptic levels, astrocytes can integrate into inhibitory neural networks to interact with neurons in neuronal circuits. Astrocytes are closely related to the pathogenesis and pathological processes of neurodegenerative diseases (NDs). Recently, optogenetics has now been applied to reveal the function of astrocytes in physiology and pathology. Herein, we discuss the possibility whether optogenetics could be used to control the release of gliotransmitters and regulate astrocytic membrane channels. Thus, the capability of modulating the bidirectional interactions between astrocytes and neurons in both synaptic and neuronal networks via optogenetics is evaluated. Furthermore, we discuss that manipulating astrocytes via optogenetics might be an effective way to investigate the potential therapeutic strategy for NDs.
Collapse
Affiliation(s)
- Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Qinghu Yang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Sciences, Beijing Institute of Technology, Beijing, China.,College of Life Sciences & Research Center for Resource Peptide Drugs, Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources, Yanan University, Yanan, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Sciences, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Sciences, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
34
|
Lee HJ, Huang KC, Mei G, Zong C, Mamaeva N, DeGrip WJ, Rothschild KJ, Cheng JX. Electronic Preresonance Stimulated Raman Scattering Imaging of Red-Shifted Proteorhodopsins: Toward Quantitation of the Membrane Potential. J Phys Chem Lett 2019; 10:4374-4381. [PMID: 31313926 DOI: 10.1021/acs.jpclett.9b01337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Voltage imaging allows mapping of the membrane potential in living cells. Yet, current intensity-based imaging approaches are limited to relative membrane potential changes, missing important information conveyed by the absolute value of the membrane voltage. This challenge arises from various factors affecting the signal intensity, such as concentration, illumination intensity, and photobleaching. Here, we demonstrate electronic preresonance hyperspectral stimulated Raman scattering (EPR-hSRS) for spectroscopic detection of the membrane voltage using a near-infrared-absorbing microbial rhodopsin expressed in E. coli. This newly developed near-infrared active microbial rhodopsin enables electronic preresonance SRS imaging at high sensitivity. By spectral profiling, we identified voltage-sensitive SRS peaks in the fingerprint region in single E. coli cells. These spectral signatures offer a new approach for quantitation of the absolute membrane voltage in living cells.
Collapse
Affiliation(s)
- Hyeon Jeong Lee
- Department of Electrical and Computer Engineering , Boston University , Boston , Massachusetts 02215 , United States
- Department of Biomedical Engineering , Boston University , Boston , Massachusetts 02215 , United States
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
| | - Kai-Chih Huang
- Department of Biomedical Engineering , Boston University , Boston , Massachusetts 02215 , United States
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
| | - Gaoxiang Mei
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
- Department of Physics , Boston University , Boston , Massachusetts 02215 , United States
| | - Cheng Zong
- Department of Electrical and Computer Engineering , Boston University , Boston , Massachusetts 02215 , United States
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
| | - Natalia Mamaeva
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
- Department of Physics , Boston University , Boston , Massachusetts 02215 , United States
| | - Willem J DeGrip
- Department of Biophysical Organic Chemistry, Leiden Institute of Chemistry , Leiden University , 2300 RA Leiden , The Netherlands
- Department of Biochemistry , Radboud University Medical School , 6500 HB Nijmegen , The Netherlands
| | - Kenneth J Rothschild
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
- Department of Physics , Boston University , Boston , Massachusetts 02215 , United States
- Department of Physiology and Biophysics , Boston University School of Medicine , Boston , Massachusetts 02218 , United States
| | - Ji-Xin Cheng
- Department of Electrical and Computer Engineering , Boston University , Boston , Massachusetts 02215 , United States
- Department of Biomedical Engineering , Boston University , Boston , Massachusetts 02215 , United States
- Photonics Center , Boston University , Boston , Massachusetts 02215 , United States
- Department of Physics , Boston University , Boston , Massachusetts 02215 , United States
| |
Collapse
|
35
|
Bostancıklıoğlu M. Optogenetic stimulation of serotonin nuclei retrieve the lost memory in Alzheimer's disease. J Cell Physiol 2019; 235:836-847. [PMID: 31332785 DOI: 10.1002/jcp.29077] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
How are memories stored and retrieved? It was one of the most discussed questions in the past century by neuroscientists. Leading studies of the period brought two different explanations to this question: The first statement considers memory as a physiological change in the brain and suggest that the retrieval of memory is only occurred by the same physiologic changes observed during the memory formation, while the second suggests that memory is a psychic mood stored in mind and the retrieval of memory is occurred by mystical energy fluctuations. Although the exact reason and the pathogenesis of Alzheimer's disease have not yet been fully understood, the approaches that centered the retrieval strategy of lost memory constitutes the basis of the treatment strategies in Alzheimer's disease today. The majority of treatment studies has based on the manipulation of the cholinergic system; however, although serotonin has mnemonic effects, its role in the pathogenesis of Alzheimer's disease has not been investigated as much as the cholinergic system. Here we show how serotonin affects the pathogenesis of Alzheimer's disease in a comprehensive perspective and we suggest that the optogenetics manipulation of serotonin nuclei retrieve the lost memory by closing the inward-rectifier potassium channel Kir2 on the memory engram cells. Also, we raise the possible effects of serotonin on the memory engram cells and the interactions between the amyloid-centric hypothesis of Alzheimer's disease and the memory engram hypothesis to explain the pathophysiology of memory loss in Alzheimer's disease.
Collapse
|
36
|
Optogenetic Stimulation Enhanced Neuronal Plasticities in Motor Recovery after Ischemic Stroke. Neural Plast 2019; 2019:5271573. [PMID: 31007684 PMCID: PMC6441501 DOI: 10.1155/2019/5271573] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/01/2019] [Accepted: 01/20/2019] [Indexed: 12/16/2022] Open
Abstract
Motor capability recovery after ischemic stroke involves dynamic remodeling processes of neural connectomes in the nervous system. Various neuromodulatory strategies combining direct stimulating interventions with behavioral trainings for motor recovery after ischemic stroke have been developed. However, the effectiveness of these interventions varies widely due to unspecific activation or inhibition of undefined neuronal subtypes. Optogenetics is a functional and structural connection-based approach that can selectively activate or inhibit specific subtype neurons with a higher precision, and it has been widely applied to build up neuronal plasticities of the nervous system, which shows a great potential in restoring motor functions in stroke animal models. Here, we reviewed neurobiological mechanisms of enhanced brain plasticities underlying motor recovery through the optogenetic stimulation after ischemic stroke. Several brain sites and neural circuits that have been previously proven effective for motor function rehabilitation were identified, which would be helpful for a more schematic understanding of effective neuronal connectomes in the motor function recovery after ischemic stroke.
Collapse
|
37
|
Anderson HE, Weir RFF. On the development of optical peripheral nerve interfaces. Neural Regen Res 2019; 14:425-436. [PMID: 30539808 PMCID: PMC6334609 DOI: 10.4103/1673-5374.245461] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 09/19/2018] [Indexed: 11/04/2022] Open
Abstract
Limb loss and spinal cord injury are two debilitating conditions that continue to grow in prevalence. Prosthetic limbs and limb reanimation present two ways of providing affected individuals with means to interact in the world. These techniques are both dependent on a robust interface with the peripheral nerve. Current methods for interfacing with the peripheral nerve tend to suffer from low specificity, high latency and insufficient robustness for a chronic implant. An optical peripheral nerve interface may solve some of these problems by decreasing invasiveness and providing single axon specificity. In order to implement such an interface three elements are required: (1) a transducer capable of translating light into a neural stimulus or translating neural activity into changes in fluorescence, (2) a means for delivering said transducer and (3) a microscope for providing the stimulus light and detecting the fluorescence change. There are continued improvements in both genetically encoded calcium and voltage indicators as well as new optogenetic actuators for stimulation. Similarly, improvements in specificity of viral vectors continue to improve expression in the axons of the peripheral nerve. Our work has recently shown that it is possible to virally transduce axons of the peripheral nerve for recording from small fibers. The improvements of these components make an optical peripheral nerve interface a rapidly approaching alternative to current methods.
Collapse
Affiliation(s)
- Hans E. Anderson
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| | - Richard F. ff. Weir
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
38
|
Mattingly M, Weineck K, Costa J, Cooper RL. Hyperpolarization by activation of halorhodopsin results in enhanced synaptic transmission: Neuromuscular junction and CNS circuit. PLoS One 2018; 13:e0200107. [PMID: 29969493 PMCID: PMC6029800 DOI: 10.1371/journal.pone.0200107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022] Open
Abstract
Optogenetics offers a unique method to regulate the activity of select neural circuits. However, the electrophysiological consequences of targeted optogenetic manipulation upon the entire circuit remain poorly understood. Analysis of the sensory-CNS-motor circuit in Drosophila larvae expressing eHpHR and ChR2-XXL revealed unexpected patterns of excitability. Optical stimulation of motor neurons targeted to express eNpHR resulted in inhibition followed by excitation of body wall contraction with repetitive stimulation in intact larvae. In situ preparations with direct electrophysiological measures showed an increased responsiveness to excitatory synaptic activity induced by sensory stimulation within a functional neural circuit. To ensure proper function of eNpHR and ChR2-XXL they were expressed in body wall muscle and direct electrophysiological measurements were obtained. Under eNpHR induced hyperpolarization the muscle remained excitable with increased amplitude of excitatory postsynaptic synaptic potentials. Theoretical models to explain the observations are presented. This study aids in increasing the understanding of the varied possible influences with light activated proteins within intact neural circuits.
Collapse
Affiliation(s)
- Matthew Mattingly
- Department of Biology and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kristin Weineck
- Department of Biology and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States of America
- Goethe University Frankfurt am Main, Frankfurt, Germany
| | - Jennifer Costa
- Department of Biology and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Robin L. Cooper
- Department of Biology and Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|