1
|
Bordett R, Danazumi KB, Wijekoon S, Garcia CJ, Abdulmalik S, Kumbar SG. Advancements in stimulation therapies for peripheral nerve regeneration. Biomed Mater 2024; 19:052008. [PMID: 39025114 PMCID: PMC11425301 DOI: 10.1088/1748-605x/ad651d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/18/2024] [Indexed: 07/20/2024]
Abstract
Soft-tissue injuries affecting muscles, nerves, vasculature, tendons, and ligaments often diminish the quality of life due to pain, loss of function, and financial burdens. Both natural healing and surgical interventions can result in scarring, which potentially may impede functional recovery and lead to persistent pain. Scar tissue, characterized by a highly disorganized fibrotic extracellular matrix, may serve as a physical barrier to regeneration and drug delivery. While approaches such as drugs, biomaterials, cells, external stimulation, and other physical forces show promise in mitigating scarring and promoting regenerative healing, their implementation remains limited and challenging. Ultrasound, laser, electrical, and magnetic forms of external stimulation have been utilized to promote soft tissue as well as neural tissue regeneration. After stimulation, neural tissues experience increased proliferation of Schwann cells, secretion of neurotropic factors, production of myelin, and growth of vasculature, all aimed at supporting axon regeneration and innervation. Yet, the outcomes of healing vary depending on the pathophysiology of the damaged nerve, the timing of stimulation following injury, and the specific parameters of stimulation employed. Increased treatment intensity and duration have been noted to hinder the healing process by inducing tissue damage. These stimulation modalities, either alone or in combination with nerve guidance conduits and scaffolds, have been demonstrated to promote healing. However, the literature currently lacks a detailed understanding of the stimulation parameters used for nerve healing applications. In this article, we aim to address this gap by summarizing existing reports and providing an overview of stimulation parameters alongside their associated healing outcomes.
Collapse
Affiliation(s)
- Rosalie Bordett
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, United States of America
| | - Khadija B Danazumi
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, United States of America
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States of America
| | - Suranji Wijekoon
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, United States of America
| | - Christopher J Garcia
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, United States of America
| | - Sama Abdulmalik
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, United States of America
| | - Sangamesh G Kumbar
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, United States of America
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States of America
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, United States of America
| |
Collapse
|
2
|
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
|
3
|
Hyung S, Park JH, Jung K. Application of optogenetic glial cells to neuron-glial communication. Front Cell Neurosci 2023; 17:1249043. [PMID: 37868193 PMCID: PMC10585272 DOI: 10.3389/fncel.2023.1249043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Optogenetic techniques combine optics and genetics to enable cell-specific targeting and precise spatiotemporal control of excitable cells, and they are increasingly being employed. One of the most significant advantages of the optogenetic approach is that it allows for the modulation of nearby cells or circuits with millisecond precision, enabling researchers to gain a better understanding of the complex nervous system. Furthermore, optogenetic neuron activation permits the regulation of information processing in the brain, including synaptic activity and transmission, and also promotes nerve structure development. However, the optimal conditions remain unclear, and further research is required to identify the types of cells that can most effectively and precisely control nerve function. Recent studies have described optogenetic glial manipulation for coordinating the reciprocal communication between neurons and glia. Optogenetically stimulated glial cells can modulate information processing in the central nervous system and provide structural support for nerve fibers in the peripheral nervous system. These advances promote the effective use of optogenetics, although further experiments are needed. This review describes the critical role of glial cells in the nervous system and reviews the optogenetic applications of several types of glial cells, as well as their significance in neuron-glia interactions. Together, it briefly discusses the therapeutic potential and feasibility of optogenetics.
Collapse
Affiliation(s)
- Sujin Hyung
- Precision Medicine Research Institute, Samsung Medical Center, Seoul, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Ji-Hye Park
- Graduate School of Cancer Science and Policy, Cancer Biomedical Science, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Kyuhwan Jung
- DAWINBIO Inc., Hanam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
4
|
Terstege DJ, Epp JR. Parvalbumin as a sex-specific target in Alzheimer's disease research - A mini-review. Neurosci Biobehav Rev 2023; 153:105370. [PMID: 37619647 DOI: 10.1016/j.neubiorev.2023.105370] [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: 05/23/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, and both the incidence of this disease and its associated cognitive decline disproportionally effect women. While the etiology of AD is unknown, recent work has demonstrated that the balance of excitatory and inhibitory activity across the brain may serve as a strong predictor of cognitive impairments in AD. Across the cortex, the most prominent source of inhibitory signalling is from a class of parvalbumin-expressing interneurons (PV+). In this mini-review, the impacts of sex- and age-related factors on the function of PV+ neurons are examined within the context of vulnerability to AD pathology. These primary factors of influence include changes in brain metabolism, circulating sex hormone levels, and inflammatory response. In addition to positing the increased vulnerability of PV+ neurons to dysfunction in AD, this mini-review highlights the critical importance of presenting sex stratified data in the study of AD.
Collapse
Affiliation(s)
- Dylan J Terstege
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
5
|
Ferreira LVDO, Kamura BDC, de Oliveira JPM, Chimenes ND, de Carvalho M, dos Santos LA, Dias-Melicio LA, Amorim RL, Amorim RM. In Vitro Transdifferentiation Potential of Equine Mesenchymal Stem Cells into Schwann-Like Cells. Stem Cells Dev 2023; 32:422-432. [PMID: 37071193 PMCID: PMC10401561 DOI: 10.1089/scd.2022.0274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/17/2023] [Indexed: 04/19/2023] Open
Abstract
Schwann cells (SCs) are essential for the regenerative processes of peripheral nerve injuries. However, their use in cell therapy is limited. In this context, several studies have demonstrated the ability of mesenchymal stem cells (MSCs) to transdifferentiate into Schwann-like cells (SLCs) using chemical protocols or co-culture with SCs. Here, we describe for the first time the in vitro transdifferentiation potential of MSCs derived from equine adipose tissue (AT) and equine bone marrow (BM) into SLCs using a practical method. In this study, the facial nerve of a horse was collected, cut into fragments, and incubated in cell culture medium for 48 h. This medium was used to transdifferentiate the MSCs into SLCs. Equine AT-MSCs and BM-MSCs were incubated with the induction medium for 5 days. After this period, the morphology, cell viability, metabolic activity, gene expression of glial markers glial fibrillary acidic protein (GFAP), myelin basic protein (MBP), p75 and S100β, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and glial cell-derived neurotrophic factor (GDNF), and the protein expression of S100 and GFAP were evaluated in undifferentiated and differentiated cells. The MSCs from the two sources incubated with the induction medium exhibited similar morphology to the SCs and maintained cell viability and metabolic activity. There was a significant increase in the gene expression of BDNF, GDNF, GFAP, MBP, p75, and S100β in equine AT-MSCs and GDNF, GFAP, MBP, p75, and S100β in equine BM-MSCs post-differentiation. Immunofluorescence analysis revealed GFAP expression in undifferentiated and differentiated cells, with a significant increase in the integrated pixel density in differentiated cells and S100 was only expressed in differentiated cells from both sources. These findings indicate that equine AT-MSCs and BM-MSCs have great transdifferentiation potential into SLCs using this method, and they represent a promising strategy for cell-based therapy for peripheral nerve regeneration in horses.
Collapse
Affiliation(s)
- Lucas Vinícius de Oliveira Ferreira
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Beatriz da Costa Kamura
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - João Pedro Marmol de Oliveira
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Natielly Dias Chimenes
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Márcio de Carvalho
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Leandro Alves dos Santos
- Confocal Microscopy Laboratory, UNIPEX–Experimental Research Unit, Medical School of Botucatu; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Luciane Alarcão Dias-Melicio
- Confocal Microscopy Laboratory, UNIPEX–Experimental Research Unit, Medical School of Botucatu; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Department of Pathology, Medical School of Botucatu; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Renée Laufer Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Rogério Martins Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Translational Nucleus of Regenerative Medicine (NUTRAMERE), School of Veterinary Medicine and Animal Science; São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
6
|
Wu Q, Xie J, Zhu X, He J. Runt-related transcription factor 3, mediated by DNA-methyltransferase 1, regulated Schwann cell proliferation and myelination during peripheral nerve regeneration via JAK/STAT signaling pathway. Neurosci Res 2023:S0168-0102(23)00008-1. [PMID: 36690210 DOI: 10.1016/j.neures.2023.01.008] [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: 09/07/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
Schwann cells (SCs) play a crucial role in peripheral nerve injury and regeneration. Recently, RUNX3 was found to be linked with neurological dysfunction. We examined the RUNX3 expression in sciatic nerve stumps with peripheral nerve injury of rats, cyclic adenosine monophosphate (cAMP)-induced SCs. MTT assay was applied to determine the proliferation of SCs. Cell migration and apoptosis were assessed using wound healing assay and flow cytometry. Subsequently, we detected the methylation level of RUNX3 using Methylation-specific PCR assay and verified its regulation by DNMT1. The RUNX3 expressions were increased in sciatic nerve stumps with peripheral nerve injury and cAMP-induced SCs differentiation, which were related to demethylation of its promoter region regulated by DNMT1. RUNX3 knockdown notably suppressed the proliferation and migration, and induced the cell apoptosis of SCs. Silencing of RUNX3 inhibited the cAMP-induced morphological changes of SCs and the increase of myelin-related proteins induced by cAMP in SCs, while RUNX3 overexpression exerted opposite effects. Besides, the overexpression of RUNX3 promoted the activation of JAK/STAT signaling to regulate SCs proliferation and myelination. Meanwhile, DNMT1 overexpression inhibited the expression of RUNX3, and cell proliferation and myelination. In conclusion, RUNX3 mediated by DNMT1 regulated SC proliferation and myelination via JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Qiufeng Wu
- Department of Neurosurgery, Xianyang Central Hospital, Xianyang, Shaanxi 712000, China
| | - Jiangtao Xie
- Department of Neurosurgery, Xianyang Central Hospital, Xianyang, Shaanxi 712000, China
| | - Xiaoli Zhu
- Department of Neurosurgery, Xianyang Central Hospital, Xianyang, Shaanxi 712000, China
| | - Juan He
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Xianyang, Xianyang, Shaanxi 712000, China.
| |
Collapse
|
7
|
Neghab HK, Djavid GE, Azadeh SS, Soheilifar MH. Osteogenic Differentiation of Menstrual Blood-Derived Stem Cells by Optogenetics. J Med Biol Eng 2022. [DOI: 10.1007/s40846-022-00714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
8
|
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: 12] [Impact Index Per Article: 3.0] [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
|
9
|
Shams Najafabadi H, Sadeghi M, Zibaii MI, Soheili ZS, Samiee S, Ghasemi P, Hosseini M, Gholami Pourbadie H, Ahmadieh H, Taghizadeh S, Ranaei Pirmardan E. Optogenetic control of neural differentiation in Opto-mGluR6 engineered retinal pigment epithelial cell line and mesenchymal stem cells. J Cell Biochem 2021; 122:851-869. [PMID: 33847009 DOI: 10.1002/jcb.29918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 11/11/2022]
Abstract
In retinal degenerative disorders, when neural retinal cells are damaged, cell transplantation is one of the most promising therapeutic approaches. Optogenetic technology plays an essential role in the neural differentiation of stem cells via membrane depolarization. This study explored the efficacy of blue light stimulation in neuroretinal differentiation of Opto-mGluR6-engineered mouse retinal pigment epithelium (mRPE) and bone marrow mesenchymal stem cells (BMSCs). mRPE and BMSCs were selected for optogenetic study due to their capability to differentiate into retinal-specific neurons. BMSCs were isolated and phenotypically characterized by the expression of mesenchymal stem cell-specific markers, CD44 (99%) and CD105 (98.8%). mRPE culture identity was confirmed by expression of RPE-specific marker, RPE65, and epithelial cell marker, ZO-1. mRPE cells and BMSCs were transduced with AAV-MCS-IRES-EGFP-Opto-mGluR6 viral vector and stimulated for 5 days with blue light (470 nm). RNA and protein expression of Opto-mGluR6 were verified. Optogenetic stimulation-induced elevated intracellular Ca2+ levels in mRPE- and BMS-treated cells. Significant increase in cell growth rate and G1/S phase transition were detected in mRPE- and BMSCs-treated cultures. Pou4f1, Dlx2, Eomes, Barlh2, Neurod2, Neurod6, Rorb, Rxrg, Nr2f2, Ascl1, Hes5, and Sox8 were overexpressed in treated BMSCs and Barlh2, Rorb, and Sox8 were overexpressed in treated mRPE cells. Expression of Rho, Thy1, OPN1MW, Recoverin, and CRABP, as retinal-specific neuron markers, in mRPE and BMS cell cultures were demonstrated. Differentiation of ganglion, amacrine, photoreceptor cells, and bipolar and Muller precursors were determined in BMSCs-treated culture and were compared with mRPE. mRPE cells represented more abundant terminal Muller glial differentiation compared with BMSCs. Our results also demonstrated that optical stimulation increased the intracellular Ca2+ level and proliferation and differentiation of Opto-mGluR6-engineered BMSCs. It seems that optogenetic stimulation of mRPE- and BMSCs-engineered cells would be a potential therapeutic approach for retinal degenerative disorders.
Collapse
Affiliation(s)
- Hoda Shams Najafabadi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehdi Sadeghi
- Department of Medical Genetics, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohammad I Zibaii
- Laser & Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Zahra-Soheila Soheili
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Shahram Samiee
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Pouria Ghasemi
- Laser & Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Hosseini
- Laser & Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| | | | - Hamid Ahmadieh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Taghizadeh
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ehsan Ranaei Pirmardan
- Molecular Biomarkers Nano-imaging Laboratory, Brigham & Women's Hospital, Department of Radiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Asano T, Teh DBL, Yawo H. Application of Optogenetics for Muscle Cells and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:359-375. [PMID: 33398826 DOI: 10.1007/978-981-15-8763-4_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This chapter describes the current progress of basic research, and potential therapeutic applications primarily focused on the optical manipulation of muscle cells and neural stem cells using microbial rhodopsin as a light-sensitive molecule. Since the contractions of skeletal, cardiac, and smooth muscle cells are mainly regulated through their membrane potential, several studies have been demonstrated to up- or downregulate the muscle contraction directly or indirectly using optogenetic actuators or silencers with defined stimulation patterns and intensities. Light-dependent oscillation of membrane potential also facilitates the maturation of myocytes with the development of T tubules and sarcomere structures, tandem arrays of minimum contractile units consists of contractile proteins and cytoskeletal proteins. Optogenetics has been applied to various stem cells and multipotent/pluripotent cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) to generate light-sensitive neurons and to facilitate neuroscience. The chronic optical stimulation of the channelrhodopsin-expressing neural stem cells facilitates their neural differentiation. There are potential therapeutic applications of optogenetics in cardiac pacemaking, muscle regeneration/maintenance, locomotion recovery for the treatment of muscle paralysis due to motor neuron diseases such as amyotrophic lateral sclerosis (ALS). Optogenetics would also facilitate maturation, network integration of grafted neurons, and improve the microenvironment around them when applied to stem cells.
Collapse
Affiliation(s)
- Toshifumi Asano
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Daniel Boon Loong Teh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiromu Yawo
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan.
| |
Collapse
|
11
|
Jung K, Kim HN, Jeon NL, Hyung S. Comparison of the Efficacy of Optogenetic Stimulation of Glia versus Neurons in Myelination. ACS Chem Neurosci 2020; 11:4280-4288. [PMID: 33269905 DOI: 10.1021/acschemneuro.0c00542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence demonstrates that optogenetics contributes to the regulation of brain behavior, cognition, and physiology, particularly during myelination, potentially allowing for the bidirectional modulation of specific cell lines with spatiotemporal accuracy. However, the type of cell to be targeted, namely, glia vs neurons, and the degree to which optogenetically induced cell activity can regulate myelination during the development of the peripheral nervous system (PNS) are still underexplored. Herein, we report the comparison of optogenetic stimulation (OS) of Schwann cells (SCs) and motor neurons (MNs) for activation of myelination in the PNS. Capitalizing on these optogenetic tools, we confirmed that the formation of the myelin sheath was initially promoted more by OS of calcium translocating channelrhodopsin (CatCh)-transfected SCs than by OS of transfected MNs at 7 days in vitro (DIV). Additionally, the level of myelination was substantially enhanced even until 14 DIV. Surprisingly, after OS of SCs, > 91.1% ± 5.9% of cells expressed myelin basic protein, while that of MNs was 67.8% ± 6.1%. The potent effect of OS of SCs was revealed by the increased thickness of the myelin sheath at 14 DIV. Thus, the OS of SCs could highly accelerate myelination, while the OS of MNs only somewhat promoted myelination, indicating a clear direction for the optogenetic application of unique cell types for initiating and promoting myelination. Together, our findings support the importance of precise cell type selection for use in optogenetics, which in turn can be broadly applied to overcome the limitations of optogenetics after injury.
Collapse
Affiliation(s)
- Kyuhwan Jung
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hong Nam Kim
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Advanced Machinery and Design, Seoul National University, Seoul 08826, Republic of Korea
| | - Sujin Hyung
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
12
|
Pezzotti G, Adachi T, Miyamoto N, Yamamoto T, Boschetto F, Marin E, Zhu W, Kanamura N, Ohgitani E, Pizzi M, Sowa Y, Mazda O. Raman Probes for In Situ Molecular Analyses of Peripheral Nerve Myelination. ACS Chem Neurosci 2020; 11:2327-2339. [PMID: 32603086 DOI: 10.1021/acschemneuro.0c00284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The myelinating activity of living Schwann cells in coculture with neuronal cells was examined in situ in a Raman microprobe spectroscope. The Raman label-free approach revealed vibrational fingerprints directly related to the activity of Schwann cells' metabolites and identified molecular species peculiar to myelinating cells. The identified chemical species included antioxidants, such as hypotaurine and glutathione, and compartmentalized water, in addition to sphingolipids, phospholipids, and nucleoside triphosphates also present in neuronal and nonmyelinating Schwann cells. Raman maps at specific frequencies could be collected, which clearly visualized the myelinating action of Schwann cells and located the demyelinated ones. An important finding was the spectroscopic visualization of confined water in the myelin structure, which exhibited a quite pronounced Raman signal at ∼3470 cm-1. This peculiar signal, whose spatial location precisely corresponded to a low-frequency fingerprint of hypotaurine, was absent in unmyelinating cells and in bulk water. Raman enhancement was attributed to frustration in the hydrogen-bond network as induced by interactions with lipids in the myelin sheaths. According to a generally accepted morphological model of myelin, an explanation was offered of the peculiar Raman scattering of water confined in intraperiod lines, according to an ordered hydrogen bonding structure. The possibility of concurrently mapping antioxidant molecules and compartmentalized water structure with high spectral accuracy and microscopic spatial resolution enables probing myelinating activity and might play a key-role in future studies of neuronal pathologies. Compatible with life, Raman microprobe spectroscopy with the newly discovered probes could be suitable for developing advanced strategies in the reconstruction of injured nerves and nerve terminals at neuromuscular junctions.
Collapse
Affiliation(s)
- Giuseppe Pezzotti
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Department of Orthopedic Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
- The Center for Advanced Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0854, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Nao Miyamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Infectious Diseases, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Francesco Boschetto
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Elia Marin
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Eriko Ohgitani
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
13
|
Zhang Z, Li X, Li A, Wu G. miR-485-5p suppresses Schwann cell proliferation and myelination by targeting cdc42 and Rac1. Exp Cell Res 2020; 388:111803. [DOI: 10.1016/j.yexcr.2019.111803] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 10/25/2022]
|
14
|
Xu X, Mee T, Jia X. New era of optogenetics: from the central to peripheral nervous system. Crit Rev Biochem Mol Biol 2020; 55:1-16. [PMID: 32070147 DOI: 10.1080/10409238.2020.1726279] [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] [Indexed: 12/15/2022]
Abstract
Optogenetics has recently gained recognition as a biological technique to control the activity of cells using light stimulation. Many studies have applied optogenetics to cell lines in the central nervous system because it has the potential to elucidate neural circuits, treat neurological diseases and promote nerve regeneration. There have been fewer studies on the application of optogenetics in the peripheral nervous system. This review introduces the basic principles and approaches of optogenetics and summarizes the physiology and mechanism of opsins and how the technology enables bidirectional control of unique cell lines with superior spatial and temporal accuracy. Further, this review explores and discusses the therapeutic potential for the development of optogenetics and its capacity to revolutionize treatment for refractory epilepsy, depression, pain, and other nervous system disorders, with a focus on neural regeneration, especially in the peripheral nervous system. Additionally, this review synthesizes the latest preclinical research on optogenetic stimulation, including studies on non-human primates, summarizes the challenges, and highlights future perspectives. The potential of optogenetic stimulation to optimize therapy for peripheral nerve injuries (PNIs) is also highlighted. Optogenetic technology has already generated exciting, preliminary evidence, supporting its role in applications to several neurological diseases, including PNIs.
Collapse
Affiliation(s)
- Xiang Xu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thomas Mee
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Hyung S, Lee S, Kim YJ, Bang S, Tahk D, Park J, Suh JF, Jeon NL. Optogenetic neuronal stimulation promotes axon outgrowth and myelination of motor neurons in a three‐dimensional motor neuron–Schwann cell coculture model on a microfluidic biochip. Biotechnol Bioeng 2019; 116:2425-2438. [DOI: 10.1002/bit.27083] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/02/2019] [Accepted: 06/06/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Sujin Hyung
- Center for BionicsKorea Institute of Science and Technology Seoul South Korea
- BK21 Plus Transformative Training Program for Creative Mechanical and Aerospace EngineersSeoul National University Seoul South Korea
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering, Institute of Advanced Machinery and DesignSeoul National University Seoul South Korea
| | - Seung‐Ryeol Lee
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering, Institute of Advanced Machinery and DesignSeoul National University Seoul South Korea
| | - Yeon Jee Kim
- Center for BionicsKorea Institute of Science and Technology Seoul South Korea
| | - Seokyoung Bang
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering, Institute of Advanced Machinery and DesignSeoul National University Seoul South Korea
| | - Dongha Tahk
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering, Institute of Advanced Machinery and DesignSeoul National University Seoul South Korea
| | - Jong‐Chul Park
- Department of Medical Engineering and Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of Medicine Seoul South Korea
| | - Jun‐Kyo Francis Suh
- Center for BionicsKorea Institute of Science and Technology Seoul South Korea
| | - Noo Li Jeon
- Multiscale Mechanical Design School of Mechanical and Aerospace Engineering, Institute of Advanced Machinery and DesignSeoul National University Seoul South Korea
| |
Collapse
|