1
|
Du L, Zhang L, Bao S, Yan F, Jiang W, Wang H, Dong C. Electric Stimulation Combined with Biomaterials for Repairing Spinal Cord Injury. ACS Biomater Sci Eng 2025. [PMID: 40403155 DOI: 10.1021/acsbiomaterials.5c00615] [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: 05/24/2025]
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) disease with a high disability rate, and reconstructing motor function after SCI remains a global challenge. Recent advancements in rehabilitation and regenerative medicine offer new approaches to SCI repair. Electrical stimulation has been shown to alter cell membrane charge distribution, generating action potentials, and affecting cell behavior. This method aids axon regeneration and neurotrophic factor upregulation, crucial for nerve repair. Biomaterials, used as scaffolds or coatings in cell culture and tissue engineering, enhance cell proliferation, migration, differentiation, and tissue regeneration. Electroactive biomaterials combined with electrical stimulation show promise in regenerating nerve, heart, and bone tissues. In this paper, different types of electrical stimulation and biomaterials applied to SCI are described, and the current application and research progress of electrical stimulation combined with biomaterials in the treatment of SCI are described, as well as the future prospects and challenges.
Collapse
Affiliation(s)
- Lulu Du
- Department of Anatomy, Medical College of Nantong University, Nantong 226019, China
| | - Liya Zhang
- Department of Anatomy, Medical College of Nantong University, Nantong 226019, China
| | - Shengzhe Bao
- Department of Anatomy, Medical College of Nantong University, Nantong 226019, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong 226019, China
| | - Wenwei Jiang
- Department of Anatomy, Medical College of Nantong University, Nantong 226019, China
| | - Hui Wang
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong 226019, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, Jiangsu Province China
| |
Collapse
|
2
|
Liu Y, Li X, Xu H, Sun K, Gong HJ, Luo C. Spinal cord stimulation induces Neurotrophin-3 to improve diabetic foot disease. Med Mol Morphol 2025; 58:43-52. [PMID: 39550735 PMCID: PMC11829938 DOI: 10.1007/s00795-024-00410-2] [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: 08/13/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024]
Abstract
Low-extremity ischemic disease is a common complication in diabetic patients, leading to reduced quality of life and potential amputation. This study investigated the therapeutic effect of spinal cord stimulation (SCS) on patients with diabetic foot disease and a rat model of diabetic foot injury. SCS was applied to patients with diabetic foot disease, with clinical assessments performed before and after therapy. Blood levels of NGF, BDNF, and NT-3 were determined by ELISA. A rat model of diabetic foot injury was established to validate NT-3's role in SCS therapy. SCS therapy improved the condition of patients with diabetic ischemic foot disease and promoted wound healing in the rat model. NT-3 levels significantly increased after SCS therapy in both patients and rats. Recombinant NT-3 administration improved wound healing and re-vascularization in the rat model, while NT-3 neutralization abrogated SCS's therapeutic effect. SCS improves the condition of patients with diabetic ischemic foot disease by inducing NT-3 production. Both SCS and NT-3 supplementation show therapeutic potential for ameliorating diabetic foot disease.
Collapse
Affiliation(s)
- Yi Liu
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - XuanPeng Li
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - HaiWen Xu
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - Ke Sun
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China
| | - Hui Jun Gong
- Department of Neurosurgery, National Regional Trauma Center, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China.
| | - Cheng Luo
- The Second Department of Neurosurgery, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming, Yunnan Province, China.
| |
Collapse
|
3
|
Liao W, Shi Y, Li Z, Yin X. Advances in 3D printing combined with tissue engineering for nerve regeneration and repair. J Nanobiotechnology 2025; 23:5. [PMID: 39754257 PMCID: PMC11697815 DOI: 10.1186/s12951-024-03052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/29/2024] [Indexed: 01/06/2025] Open
Abstract
The repair of nerve damage has long posed a challenge owing to limited self-repair capacity and the highly differentiated nature of nerves. While new therapeutic and pharmacologic interventions have emerged in neurology, their regenerative efficacy remains limited. Tissue engineering offers a promising avenue for overcoming the limitations of conventional treatments and increasing the outcomes of regenerative repair. By implanting scaffolds into damaged nerve tissue sites, the repair and functional reconstruction of nerve injuries can be significantly facilitated. The integration of three-dimensional (3D) printing technology introduces a novel approach for accurate simulation and scalably fabricating neural tissue structures. Tissue-engineered scaffolds developed through 3D printing technology are expected to be a viable therapeutic option for nerve injuries, with broad applicability and continued development. This review systematically examines recent advances in 3D printing and tissue engineering for nerve regeneration and repair. It details the basic principles and construction strategies of neural tissue engineering and explores the crucial role of 3D printing technology. Additionally, it elucidates specific applications and technical challenges associated with this integrated approach, thereby providing valuable insights into innovative strategies and pragmatic implementation within this field.
Collapse
Affiliation(s)
- Weifang Liao
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
| | - Yuying Shi
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China
| | - Zuguang Li
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, No. 57 East Xunyang Road, Jiujiang, Jiangxi, 332005, China.
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, China.
| |
Collapse
|
4
|
Li WY, Qu WR, Li Y, Wang SY, Liu DM, Deng LX, Wang Y. DBS in the restoration of motor functional recovery following spinal cord injury. Front Neurol 2024; 15:1442281. [PMID: 39697443 PMCID: PMC11652279 DOI: 10.3389/fneur.2024.1442281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
The landscape of therapeutic deep brain stimulation (DBS) for locomotor function recovery is rapidly evolving. This review provides an overview of electrical neuromodulation effects on spinal cord injury (SCI), focusing on DBS for motor functional recovery in human and animal models. We highlight research providing insight into underlying cellular and molecular mechanisms. A literature review via Web of Science and PubMed databases from 1990 to May 29, 2024, reveals a growing body of evidence for therapeutic DBS in SCI recovery. Advances in techniques like optogenetics and whole-brain tractogram have helped elucidate DBS mechanisms. Neuronal targets sites for SCI functional recovery include the mesencephalic locomotor region (MLR), cuneiform nucleus (CNF), and nucleus raphe magnus (NRG), with pedunculopontine nucleus (PPN), periaqueductal gray (PAG), and nucleus ventroposterolateral thalami (VPL) for post-injury functional recovery treatment. Radiologically guided DBS optimization and combination therapy with classical rehabilitation have become an effective therapeutic method, though ongoing interventional trials are needed to enhance understanding and validate DBS efficacy in SCI. On the pre-clinical front, standardization of pre-clinical approaches are essential to enhance the quality of evidence on DBS safety and efficacy. Mapping brain targets and optimizing DBS protocols, aided by combined DBS and medical imaging, are critical endeavors. Overall, DBS holds promise for neurological and functional recovery after SCI, akin to other electrical stimulation approaches.
Collapse
Affiliation(s)
- Wen-yuan Li
- Mudanjiang North Medicine Resource Development and Application Collaborative Innovation Center, Mudanjiang, China
- Institute of Neural Tissue Engineering, Mudanjiang University of Medicine, Mudanjiang, China
| | - Wen-rui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yi Li
- Mudanjiang North Medicine Resource Development and Application Collaborative Innovation Center, Mudanjiang, China
- Institute of Neural Tissue Engineering, Mudanjiang University of Medicine, Mudanjiang, China
| | - Shu-ying Wang
- Mudanjiang North Medicine Resource Development and Application Collaborative Innovation Center, Mudanjiang, China
- Institute of Neural Tissue Engineering, Mudanjiang University of Medicine, Mudanjiang, China
| | - Dong-ming Liu
- Department of Neurology, Mudanjiang First People’s Hospital, Mudanjiang, China
| | - Ling-xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ying Wang
- Mudanjiang North Medicine Resource Development and Application Collaborative Innovation Center, Mudanjiang, China
- Institute of Neural Tissue Engineering, Mudanjiang University of Medicine, Mudanjiang, China
| |
Collapse
|
5
|
Hardy PB, Wang BY, Chan KM, Webber CA, Senger JLB. The use of electrical stimulation to enhance recovery following peripheral nerve injury. Muscle Nerve 2024; 70:1151-1162. [PMID: 39347555 DOI: 10.1002/mus.28262] [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: 01/17/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024]
Abstract
Peripheral nerve injury is common and can have devastating consequences. In severe cases, functional recovery is often poor despite surgery. This is primarily due to the exceedingly slow rate of nerve regeneration at only 1-3 mm/day. The local environment in the distal nerve stump supportive of nerve regrowth deteriorates over time and the target end organs become atrophic. To overcome these challenges, investigations into treatments capable of accelerating nerve regrowth are of great clinical relevance and are an active area of research. One intervention that has shown great promise is perioperative electrical stimulation. Postoperative stimulation helps to expedite the Wallerian degeneration process and reduces delays caused by staggered regeneration at the site of nerve injury. By contrast, preoperative "conditioning" stimulation increases the rate of nerve regrowth along the nerve trunk. Over the past two decades, a rich body of literature has emerged that provides molecular insights into the mechanism by which electrical stimulation impacts nerve regeneration. The end result is upregulation of regeneration-associated genes in the neuronal body and accelerated transport to the axon front for regrowth. The efficacy of brief electrical stimulation on patients with peripheral nerve injuries was demonstrated in a number of randomized controlled trials on compressive, transection and traction injuries. As approved equipment to deliver this treatment is becoming available, it may be feasible to deploy this novel treatment in a wide range of clinical settings.
Collapse
Affiliation(s)
- Paige B Hardy
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Bonnie Y Wang
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Alberta, Canada
| | - K Ming Chan
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Alberta, Canada
| | | | - Jenna-Lynn B Senger
- Division of Plastic & Reconstructive Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
6
|
Guo J, Cao J, Wu J, Gao J. Electrical stimulation and conductive materials: electrophysiology-based treatment for spinal cord injury. Biomater Sci 2024; 12:5704-5721. [PMID: 39403758 DOI: 10.1039/d4bm00959b] [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: 11/06/2024]
Abstract
Spinal cord injury is a serious disease of the central nervous system. The electrophysiological properties of the spinal cord that are essential to maintaining neurotransmission can be impaired after the injury. Therefore, electrophysiological evaluation is becoming an important indicator of the injury extent or the therapeutic outcomes by reflecting the potential propagation of neural pathways. On the other hand, the repair of damaged nerves is one of the main goals of spinal cord injury treatment. Growing research interest has been concentrated on developing effective therapeutic solutions to restore the normal electrophysiological function of the injured spinal cord by using conductive materials and/or exerting the merits of electrical stimulation. Accordingly, this review introduces the current common electrophysiological evaluation in spinal cord injury. Then the cutting-edge therapeutic strategies aiming at electrophysiological improvement in spinal cord injury are summarized. Finally, the challenges and future prospects of neural restoration after spinal cord injury are presented.
Collapse
Affiliation(s)
- Jing Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jian Cao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jiahe Wu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Pharmacy, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310006, China.
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321000, China
| |
Collapse
|
7
|
Nowakowska M, Jakešová M, Schmidt T, Opančar A, Polz M, Reimer R, Fuchs J, Patz S, Ziesel D, Scheruebel S, Kornmueller K, Rienmüller T, Đerek V, Głowacki ED, Schindl R, Üçal M. Light-Controlled Electric Stimulation with Organic Electrolytic Photocapacitors Achieves Complex Neuronal Network Activation: Semi-Chronic Study in Cortical Cell Culture and Rat Model. Adv Healthc Mater 2024; 13:e2401303. [PMID: 39139004 PMCID: PMC11582505 DOI: 10.1002/adhm.202401303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Neurostimulation employing photoactive organic semiconductors offers an appealing alternative to conventional techniques, enabling targeted action and wireless control through light. In this study, organic electrolytic photocapacitors (OEPC) are employed to investigate the effects of light-controlled electric stimulation on neuronal networks in vitro and in vivo. The interactions between the devices and biological systems are characterized. Stimulation of primary rat cortical neurons results in an elevated expression of c-Fos within a mature neuronal network. OEPC implantation for three weeks and subsequent stimulation of the somatosensory cortex leads to an increase of c-Fos in neurons at the stimulation site and in connected brain regions (entorhinal cortex, hippocampus), both in the ipsi- and contralateral hemispheres. Reactivity of glial and immune cells after semi-chronic implantation of OEPC in the rat brain is comparable to that of surgical controls, indicating minimal foreign body response. Device functionality is further substantiated through retained charging dynamics following explantation. OEPC-based, light-controlled electric stimulation has a significant impact on neural responsiveness. The absence of detrimental effects on both the brain and device encourages further use of OEPC as cortical implants. These findings highlight its potential as a novel mode of neurostimulation and instigate further exploration into applications in fundamental neuroscience.
Collapse
Affiliation(s)
- Marta Nowakowska
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, Graz, 8036, Austria
- BioTechMed-Graz, Mozartgasse 12/II, Graz, 8010, Austria
| | - Marie Jakešová
- Bioelectronics Materials and Devices Laboratory, CEITEC, Brno University of Technology, Purkyňova 123, Brno, 612 00, Czech Republic
| | - Tony Schmidt
- BioTechMed-Graz, Mozartgasse 12/II, Graz, 8010, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Aleksandar Opančar
- Bioelectronics Materials and Devices Laboratory, CEITEC, Brno University of Technology, Purkyňova 123, Brno, 612 00, Czech Republic
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička c. 32, Zagreb, 10000, Croatia
| | - Mathias Polz
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Stremayrgasse 16/II, Graz, 8010, Austria
| | - Robert Reimer
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Julia Fuchs
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Stremayrgasse 16/II, Graz, 8010, Austria
| | - Silke Patz
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, Graz, 8036, Austria
| | - Daniel Ziesel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Stremayrgasse 16/II, Graz, 8010, Austria
| | - Susanne Scheruebel
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Karin Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Theresa Rienmüller
- BioTechMed-Graz, Mozartgasse 12/II, Graz, 8010, Austria
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Stremayrgasse 16/II, Graz, 8010, Austria
| | - Vedran Đerek
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička c. 32, Zagreb, 10000, Croatia
| | - Eric D Głowacki
- Bioelectronics Materials and Devices Laboratory, CEITEC, Brno University of Technology, Purkyňova 123, Brno, 612 00, Czech Republic
| | - Rainer Schindl
- BioTechMed-Graz, Mozartgasse 12/II, Graz, 8010, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6, Graz, 8010, Austria
| | - Muammer Üçal
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, Graz, 8036, Austria
- BioTechMed-Graz, Mozartgasse 12/II, Graz, 8010, Austria
- Department of Neurology, Medical University of Graz, Auenbruggerplatz 22, Graz, 8036, Austria
| |
Collapse
|
8
|
Naseri S, Samaram H, Naghavi N, Rassouli MB, Mousavinezhad M. Types of Short-Duration Electrical Stimulation-Induced Efficiency in the Axonal Regeneration and Recovery: Comparative in Vivo Study in Rat Model of Repaired Sciatic Nerve and its Tibial Branch after Transection Injury. Neurochem Res 2024; 49:2469-2479. [PMID: 38856888 DOI: 10.1007/s11064-024-04154-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/25/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
The restoration of adequate function and sensation in nerves following an injury is often insufficient. Electrical stimulation (ES) applied during nerve repair can promote axon regeneration, which may enhance the likelihood of successful functional recovery. However, increasing operation time and complexity are associated with limited clinical use of ES. This study aims to better assess whether short-duration ES types (voltage mode vs. current mode) are able to produce enhanced regenerative activity following peripheral nerve repair in rat models. Wistar rats were randomly divided into 3 groups: no ES (control), 30-minute ES with a current pulse, and 30-minute ES with a voltage pulse. All groups underwent sciatic nerve transection and repair using a silicone tube to bridge the 6-mm gap between the stumps. In the 2 groups other than the control, ES was applied after the surgical repair. Outcomes were evaluated using electrophysiology, histology, and serial walking track analysis. Biweekly walking tracks test over 12 weeks revealed that subjects that underwent ES experienced more rapid functional improvement than subjects that underwent repair alone. Electrophysiological analysis of the newly intratubular sciatic nerve at week 12 revealed strong motor function recovery in rats that underwent 30-minute ES. Histologic analysis of the sciatic nerve and its tibial branch at 12 weeks demonstrated robust axon regrowth in all groups. Both types of short-duration ES applied during nerve repair can promote axon regrowth and enhance the chances of successful functional recovery.
Collapse
Affiliation(s)
- Sareh Naseri
- Electrical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Razavi Khorasan Province, 9177948374, Iran
| | - Hosein Samaram
- Electrical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Razavi Khorasan Province, 9177948374, Iran
| | - Nadia Naghavi
- Electrical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Razavi Khorasan Province, 9177948374, Iran.
| | | | - Maryam Mousavinezhad
- Biology Department, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
9
|
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
|
10
|
Abdel-Kareem NM, Elshazly SM, Abd El Fattah MA, Aldahish AA, Zaitone SA, Ali SK, Abd El-Haleim EA. Nifedipine Improves the Ketogenic Diet Effect on Insulin-Resistance-Induced Cognitive Dysfunction in Rats. Pharmaceuticals (Basel) 2024; 17:1054. [PMID: 39204160 PMCID: PMC11359371 DOI: 10.3390/ph17081054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Insulin resistance, induced by high fructose consumption, affects cognitive function negatively. Nifedipine may be suggested for neurological disorders. This study aimed to assess the effect of nifedipine with either a normal diet (ND) or a ketogenic diet (KD) in cognitive dysfunction. Male Wistar rats received 10% fructose in drinking water for 8 weeks to induce insulin resistance. Rats received nifedipine (5.2 mg/kg/day; p.o.) later with ND or KD for an additional five weeks. One and two-way ANOVAs were used in analyzing the data. Reversion to the ND improved insulin resistance and lipid profile, besides brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3 beta (GSK3β), and insulin-degrading enzyme (IDE) levels. Rats fed KD alone and those that received nifedipine with KD did not show similar improvement in the previously mentioned parameters as the ND group. However, nifedipine-ND rats showed improvement in cognitive behavior and insulin resistance. Treatment with nifedipine-KD ameliorated GSK3β, amyloid β (Aβ), and tau protein levels. As the nifedipine-KD combination succeeded in diminishing the accumulated Aβ and tau protein, KD may be used for a while due to its side effects, then nifedipine treatment could be continued with an ND. This conclusion is based on the finding that this combination mitigated insulin resistance with the associated improved behavior.
Collapse
Affiliation(s)
- Nancy M. Abdel-Kareem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University—Arish Branch, Arish 45511, Egypt
| | - Shimaa M. Elshazly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - May A. Abd El Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt (E.A.A.E.-H.)
| | - Afaf A. Aldahish
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Sawsan A. Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Sahar K. Ali
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Enas A. Abd El-Haleim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt (E.A.A.E.-H.)
| |
Collapse
|
11
|
Senanayake J, Mattingly RR, Sundararaghavan HG. Electrical stimulation of Schwann cells on electrospun hyaluronic acid carbon nanotube fibers. PLoS One 2024; 19:e0308207. [PMID: 39110684 PMCID: PMC11305570 DOI: 10.1371/journal.pone.0308207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
Neurofibromatosis Type 1 (NF1) is a complex genetic disorder characterized by the development of benign neurofibromas, which can cause significant morbidity in affected individuals. While the molecular mechanisms underlying NF1 pathogenesis have been extensively studied, the development of effective therapeutic strategies remains a challenge. This paper presents the development and validation of a novel biomaterial testing model to enhance our understanding of NF1 pathophysiology, disease mechanisms and evaluate potential therapeutic interventions. Our long-term goal is to develop an invitro model of NF1 to evaluate drug targets. We have developed an in vitro system to test the cellular behavior of NF1 patient derived cells on electroconductive aligned nanofibrous biomaterials with electrical stimulatory cues. We hypothesized that cells cultured on electroconductive biomaterial will undergo morphological changes and variations in cell proliferation that could be further enhanced with the combination of exogenous electrical stimulation (ES). In this study, we developed electrospun Hyaluronic Acid-Carbon Nanotube (HA-CNT) nanofiber scaffolds to mimic the axon's topographical and bioelectrical cues that influence neurofibroma growth and development. The cellular behavior was qualitatively and quantitively analyzed through immunofluorescent stains, Alamar blue assays and ELISA assays. Schwann cells from NF1 patients appear to have lost their ability to respond to electrical stimulation in the development and regeneration range, which was seen through changes in morphology, proliferation and NGF release. Without stimulation, the conductive material enhances NF1 SC behavior. Wild-type SC respond to electrical stimulation with increased cell proliferation and NGF release. Using this system, we can better understand the interaction between axons and SC that lead to tumor formation, homeostasis and regeneration.
Collapse
Affiliation(s)
- Judy Senanayake
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States of America
| | - Raymond R. Mattingly
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, United States of America
| | | |
Collapse
|
12
|
Senger JL, Power H, Moore AM. Electrical Stimulation: How It Works and How to Apply It. Hand Clin 2024; 40:409-420. [PMID: 38972685 DOI: 10.1016/j.hcl.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Electrical stimulation is emerging as a perioperative strategy to improve peripheral nerve regeneration and enhance functional recovery. Despite decades of research, new insights into the complex multifaceted mechanisms of electrical stimulation continue to emerge, providing greater understanding of the neurophysiology of nerve regeneration. In this study, we summarize what is known about how electrical stimulation modulates the molecular cascades and cellular responses innate to nerve injury and repair, and the consequential effects on axonal growth and plasticity. Further, we discuss how electrical stimulation is delivered in preclinical and clinical studies and identify knowledge gaps that may provide opportunities for optimization.
Collapse
Affiliation(s)
- Jenna-Lynn Senger
- Division of Plastic Surgery, University of British Columbia, Suite 1788, 1111 W Georgia Street, Vancouver, British Columbia, V6E 4M3, Canada
| | - Hollie Power
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Suite 401, 316 Windermere Road NorthWest, Edmonton, Alberta T6W 2Z8, Canada
| | - Amy M Moore
- Department of Plastic and Reconstructive Surgery, The Ohio State University, 915 Olentangy River Road Suite 2100, Columbus, OH 43212, USA.
| |
Collapse
|
13
|
Sun Y, Xiao Z, Chen B, Zhao Y, Dai J. Advances in Material-Assisted Electromagnetic Neural Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400346. [PMID: 38594598 DOI: 10.1002/adma.202400346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Indexed: 04/11/2024]
Abstract
Bioelectricity plays a crucial role in organisms, being closely connected to neural activity and physiological processes. Disruptions in the nervous system can lead to chaotic ionic currents at the injured site, causing disturbances in the local cellular microenvironment, impairing biological pathways, and resulting in a loss of neural functions. Electromagnetic stimulation has the ability to generate internal currents, which can be utilized to counter tissue damage and aid in the restoration of movement in paralyzed limbs. By incorporating implanted materials, electromagnetic stimulation can be targeted more accurately, thereby significantly improving the effectiveness and safety of such interventions. Currently, there have been significant advancements in the development of numerous promising electromagnetic stimulation strategies with diverse materials. This review provides a comprehensive summary of the fundamental theories, neural stimulation modulating materials, material application strategies, and pre-clinical therapeutic effects associated with electromagnetic stimulation for neural repair. It offers a thorough analysis of current techniques that employ materials to enhance electromagnetic stimulation, as well as potential therapeutic strategies for future applications.
Collapse
Affiliation(s)
- Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
14
|
Albin B, Adhikari P, Tiwari AP, Qubbaj K, Yang IH. Electrical stimulation enhances mitochondrial trafficking as a neuroprotective mechanism against chemotherapy-induced peripheral neuropathy. iScience 2024; 27:109052. [PMID: 38375222 PMCID: PMC10875116 DOI: 10.1016/j.isci.2024.109052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Electrical stimulation (ESTIM) has shown to be an effective symptomatic treatment to treat pain associated with peripheral nerve damage. However, the neuroprotective mechanism of ESTIM on peripheral neuropathies is still unknown. In this study, we identified that ESTIM has the ability to enhance mitochondrial trafficking as a neuroprotective mechanism against chemotherapy-induced peripheral neuropathies (CIPNs). CIPN is a debilitating and painful sequalae of anti-cancer chemotherapy treatment which results in degeneration of peripheral nerves. Mitochondrial dynamics were analyzed within axons in response to two different antineoplastic mechanisms by chemotherapy drug treatments paclitaxel and oxaliplatin in vitro. Mitochondrial trafficking response to chemotherapy drug treatment was observed to decrease in conjunction with degeneration of distal axons. Using low-frequency ESTIM, we observed enhanced mitochondrial trafficking to be a neuroprotective mechanism against CIPN. This study confirms ESTIM enhances regeneration of peripheral nerves by increased mitochondrial trafficking.
Collapse
Affiliation(s)
- Bayne Albin
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Prashant Adhikari
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Arjun Prasad Tiwari
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Khayzaran Qubbaj
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - In Hong Yang
- Center for Biomedical Engineering and Science, Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| |
Collapse
|
15
|
Wong EC, Lopez IA, Ishiyama A, Ishiyama G. Expression of Brain-Derived Neurotrophic Factor in Human Spiral Ganglia Neurons after Cochlear Implantation. Otol Neurotol 2024; 45:326-333. [PMID: 38238917 PMCID: PMC10922350 DOI: 10.1097/mao.0000000000004104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is an important factor in the development and neuroprotection of afferent auditory pathways. In this study, we investigated the expression of BDNF in the afferent auditory pathway after cochlear implantation (CI), hypothesizing that electrical stimulation after CI stimulates BDNF expression in the afferent auditory pathway. METHODS Archival human temporal bones from eight patients with a history of CI and five patients with normal hearing (ages 65-93 years old) were studied. Temporal bone specimens were immunoreacted with rabbit polyclonal antibodies against BDNF and mouse monoclonal antibodies against pan-neurofilaments. In cases of unilateral CI, the BDNF expression was compared with the contralateral unimplanted ear and normal temporal bones without hearing loss. RESULTS BDNF immunoreactivity (IR) localized to the spiral ganglion neurons (SGNs) somata and the surrounding satellite cells. BDNF-IR in the spiral ganglia was similar in the apical, middle, and basal hook regions. Neurofilament IR localized to SGN nerve fibers in both implanted and unimplanted cochleae. BDNF-IR in the SGN and satellite cells was significantly increased in the implanted specimens compared with the unimplanted specimens ( p < 0.05) and the normal hearing specimens ( p < 0.05). BDNF-IR expression was similar in the unimplanted cochlea and in the normal cochlea. BDNF protein expression was increased despite complete loss of the organ of Corti hair cells and supporting cells. Even in the cases of CI with a 6-mm first-generation electrode, BDNF expression was upregulated throughout the cochlea. CONCLUSIONS BDNF expression in the SGN appears to be upregulated by the electrical stimulation from CI. This study provides evidence that the electrical stimulation from CI may stimulate the expression of BDNF, playing a neuroprotective role in the rehabilitation of hearing in the deafened ear.
Collapse
Affiliation(s)
| | | | | | - Gail Ishiyama
- UCLA Department of Head and Neck Surgery
- UCLA Department of Neurology
| |
Collapse
|
16
|
Sha B, Du Z. Neural repair and regeneration interfaces: a comprehensive review. Biomed Mater 2024; 19:022002. [PMID: 38232383 DOI: 10.1088/1748-605x/ad1f78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/17/2024] [Indexed: 01/19/2024]
Abstract
Neural interfaces play a pivotal role in neuromodulation, as they enable precise intervention into aberrant neural activity and facilitate recovery from neural injuries and resultant functional impairments by modulating local immune responses and neural circuits. This review outlines the development and applications of these interfaces and highlights the advantages of employing neural interfaces for neural stimulation and repair, including accurate targeting of specific neural populations, real-time monitoring and control of neural activity, reduced invasiveness, and personalized treatment strategies. Ongoing research aims to enhance the biocompatibility, stability, and functionality of these interfaces, ultimately augmenting their therapeutic potential for various neurological disorders. The review focuses on electrophysiological and optophysiology neural interfaces, discussing functionalization and power supply approaches. By summarizing the techniques, materials, and methods employed in this field, this review aims to provide a comprehensive understanding of the potential applications and future directions for neural repair and regeneration devices.
Collapse
Affiliation(s)
- Baoning Sha
- Brain Cognition and Brain Disease Institute, CAS Key Laboratory of Brain Connectome and Manipulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, People's Republic of China
- Department of Biomedical Engineering, Columbia University, New York, NY, United States of America
| | - Zhanhong Du
- Brain Cognition and Brain Disease Institute, CAS Key Laboratory of Brain Connectome and Manipulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| |
Collapse
|
17
|
Deng C, Chen H. Brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling in spinal muscular atrophy and amyotrophic lateral sclerosis. Neurobiol Dis 2024; 190:106377. [PMID: 38092270 DOI: 10.1016/j.nbd.2023.106377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023] Open
Abstract
Tropomyosin receptor kinase B (TrkB) and its primary ligand brain-derived neurotrophic factor (BDNF) are expressed in the neuromuscular system, where they affect neuronal survival, differentiation, and functions. Changes in BDNF levels and full-length TrkB (TrkB-FL) signaling have been revealed in spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS), two common forms of motor neuron diseases that are characterized by defective neuromuscular junctions in early disease stages and subsequently progressive muscle weakness. This review summarizes the current understanding of BDNF/TrkB-FL-related research in SMA and ALS, with an emphasis on their alterations in the neuromuscular system and possible BDNF/TrkB-FL-targeting therapeutic strategies. The limitations of current studies and future directions are also discussed, giving the hope of discovering novel and effective treatments.
Collapse
Affiliation(s)
- Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Qin C, Qi Z, Pan S, Xia P, Kong W, Sun B, Du H, Zhang R, Zhu L, Zhou D, Yang X. Advances in Conductive Hydrogel for Spinal Cord Injury Repair and Regeneration. Int J Nanomedicine 2023; 18:7305-7333. [PMID: 38084124 PMCID: PMC10710813 DOI: 10.2147/ijn.s436111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Spinal cord injury (SCI) treatment represents a major challenge in clinical practice. In recent years, the rapid development of neural tissue engineering technology has provided a new therapeutic approach for spinal cord injury repair. Implanting functionalized electroconductive hydrogels (ECH) in the injury area has been shown to promote axonal regeneration and facilitate the generation of neuronal circuits by reshaping the microenvironment of SCI. ECH not only facilitate intercellular electrical signaling but, when combined with electrical stimulation, enable the transmission of electrical signals to electroactive tissue and activate bioelectric signaling pathways, thereby promoting neural tissue repair. Therefore, the implantation of ECH into damaged tissues can effectively restore physiological functions related to electrical conduction. This article focuses on the dynamic pathophysiological changes in the SCI microenvironment and discusses the mechanisms of electrical stimulation/signal in the process of SCI repair. By examining electrical activity during nerve repair, we provide insights into the mechanisms behind electrical stimulation and signaling during SCI repair. We classify conductive biomaterials, and offer an overview of the current applications and research progress of conductive hydrogels in spinal cord repair and regeneration, aiming to provide a reference for future explorations and developments in spinal cord regeneration strategies.
Collapse
Affiliation(s)
- Cheng Qin
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Zhiping Qi
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Su Pan
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Peng Xia
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Weijian Kong
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Bin Sun
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Haorui Du
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Renfeng Zhang
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Longchuan Zhu
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Dinghai Zhou
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery, the Second Hospital of Jilin University, Changchun, 130041, People’s Republic of China
| |
Collapse
|
19
|
Sun P, Guan Y, Yang C, Hou H, Liu S, Yang B, Li X, Chen S, Wang L, Wang H, Huang Y, Sheng X, Peng J, Xiong W, Wang Y, Yin L. A Bioresorbable and Conductive Scaffold Integrating Silicon Membranes for Peripheral Nerve Regeneration. Adv Healthc Mater 2023; 12:e2301859. [PMID: 37750601 DOI: 10.1002/adhm.202301859] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/03/2023] [Indexed: 09/27/2023]
Abstract
Peripheral nerve injury represents one of the most common types of traumatic damage, severely impairing motor and sensory functions, and posttraumatic nerve regeneration remains a major challenge. Electrical cues are critical bioactive factors that promote nerve regrowth, and bioartificial scaffolds incorporating conductive materials to enhance the endogenous electrical field have been demonstrated to be effective. The utilization of fully biodegradable scaffolds can eliminate material residues, and circumvent the need for secondary retrieval procedures. Here, a fully bioresorbable and conductive nerve scaffold integrating N-type silicon (Si) membranes is proposed, which can deliver both structural guidance and electrical cues for the repair of nerve defects. The entire scaffold is fully biodegradable, and the introduction of N-type Si can significantly promote the proliferation and production of neurotrophic factors of Schwann cells and enhance the calcium activity of dorsal root ganglion (DRG) neurons. The conductive scaffolds enable accelerated nerve regeneration and motor functional recovery in rodents with sciatic nerve transection injuries. This work sheds light on the advancement of bioresorbable and electrically active materials to achieve desirable neural interfaces and improved therapeutic outcomes, offering essential strategies for regenerative medicine.
Collapse
Affiliation(s)
- Pengcheng Sun
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, P. R. China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Nantong, Jiangsu Province, 226007, P. R. China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, P. R. China
| | - Can Yang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| | - Hanqing Hou
- School of Life Sciences, IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing, 100084, P. R. China
| | - Shuang Liu
- School of Life Sciences, IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing, 100084, P. R. China
| | - Boyao Yang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, P. R. China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, P. R. China
| | - Xiangling Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, P. R. China
| | - Shengfeng Chen
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, P. R. China
| | - Liu Wang
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, P. R. China
| | - Huachun Wang
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Yunxiang Huang
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, P. R. China
| | - Wei Xiong
- School of Life Sciences, IDG/McGovern Institute for Brain Research at Tsinghua University, Tsinghua University, Beijing, 100084, P. R. China
| | - Yu Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, P. R. China
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
20
|
Zhong H, Xing C, Zhou M, Jia Z, Liu S, Zhu S, Li B, Yang H, Ma H, Wang L, Zhu R, Qu Z, Ning G. Alternating current stimulation promotes neurite outgrowth and plasticity in neurons through activation of the PI3K/AKT signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1718-1729. [PMID: 37814815 PMCID: PMC10679878 DOI: 10.3724/abbs.2023238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/04/2023] [Indexed: 10/11/2023] Open
Abstract
As a commonly used physical intervention, electrical stimulation (ES) has been demonstrated to be effective in the treatment of central nervous system disorders. Currently, researchers are studying the effects of electrical stimulation on individual neurons and neural networks, which are dependent on factors such as stimulation intensity, duration, location, and neuronal properties. However, the exact mechanism of action of electrical stimulation remains unclear. In some cases, repeated or prolonged electrical stimulation can lead to changes in the morphology or function of the neuron. In this study, immunofluorescence staining and Sholl analysis are used to assess changes in the neurite number and axon length to determine the optimal pattern and stimulation parameters of ES for neurons. Neuronal death and plasticity are detected by TUNEL staining and microelectrode array assays, respectively. mRNA sequencing and bioinformatics analysis are applied to predict the key targets of the action of ES on neurons, and the identified targets are validated by western blot analysis and qRT-PCR. The effects of alternating current stimulation (ACS) on neurons are more significant than those of direct current stimulation (DCS), and the optimal parameters are 3 μA and 20 min. ACS stimulation significantly increases the number of neurites, the length of axons and the spontaneous electrical activity of neurons, significantly elevates the expression of growth-associated protein-43 (GAP-43) without significant changes in the expression of neurotrophic factors. Furthermore, application of PI3K/AKT-specific inhibitors significantly abolishes the beneficial effects of ACS on neurons, confirming that the PI3K/AKT pathway is an important potential signaling pathway in the action of ACS.
Collapse
Affiliation(s)
- Hao Zhong
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Cong Xing
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Mi Zhou
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Zeyu Jia
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Song Liu
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Shibo Zhu
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Bo Li
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Hongjiang Yang
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Hongpeng Ma
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Liyue Wang
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| | - Rusen Zhu
- Department of Spine SurgeryTianjin Union Medical CenterTianjin300121China
| | - Zhigang Qu
- College of Electronic Information and AutomationAdvanced Structural Integrity International Joint Research CenterTianjin University of Science and TechnologyTianjin300222China
| | - Guangzhi Ning
- International Science and Technology Cooperation Base of Spinal Cord InjuryTianjin Key Laboratory of Spine and Spinal Cord InjuryDepartment of OrthopedicsTianjin Medical University General HospitalTianjin300052China
| |
Collapse
|
21
|
Yang Y, Rao C, Yin T, Wang S, Shi H, Yan X, Zhang L, Meng X, Gu W, Du Y, Hong F. Application and underlying mechanism of acupuncture for the nerve repair after peripheral nerve injury: remodeling of nerve system. Front Cell Neurosci 2023; 17:1253438. [PMID: 37941605 PMCID: PMC10627933 DOI: 10.3389/fncel.2023.1253438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Peripheral nerve injury (PNI) is a structural event with harmful consequences worldwide. Due to the limited intrinsic regenerative capacity of the peripheral nerve in adults, neural restoration after PNI is difficult. Neurological remodeling has a crucial effect on the repair of the form and function during the regeneration of the peripheral nerve after the peripheral nerve is injured. Several studies have demonstrated that acupuncture is effective for PNI-induced neurologic deficits, and the potential mechanisms responsible for its effects involve the nervous system remodeling in the process of nerve repair. Moreover, acupuncture promotes neural regeneration and axon sprouting by activating related neurotrophins retrograde transport, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), glial cell-derived neurotrophic factor (GDNF), N-cadherin, and MicroRNAs. Peripheral nerve injury enhances the perceptual response of the central nervous system to pain, causing central sensitization and accelerating neuronal cell apoptosis. Together with this, the remodeling of synaptic transmission function would worsen pain discomfort. Neuroimaging studies have shown remodeling changes in both gray and white matter after peripheral nerve injury. Acupuncture not only reverses the poor remodeling of the nervous system but also stimulates the release of neurotrophic substances such as nerve growth factors in the nervous system to ameliorate pain and promote the regeneration and repair of nerve fibers. In conclusion, the neurological remodeling at the peripheral and central levels in the process of acupuncture treatment accelerates nerve regeneration and repair. These findings provide novel insights enabling the clinical application of acupuncture in the treatment of PNI.
Collapse
Affiliation(s)
- Yongke Yang
- Beilun District People’s Hospital, Ningbo, China
| | - Chang Rao
- Tianjin Union Medical Center, Tianjin, China
| | - Tianlong Yin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shaokang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huiyan Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xin Yan
- National Anti-Drug Laboratory Beijing Regional Center, Beijing, China
| | - Lili Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianggang Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenlong Gu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Hong
- Beilun District People’s Hospital, Ningbo, China
| |
Collapse
|
22
|
Foo VHX, Liu YC, Tho B, Tong L. Quantum molecular resonance electrotherapy (Rexon-Eye) for recalcitrant dry eye in an Asian population. Front Med (Lausanne) 2023; 10:1209886. [PMID: 37771976 PMCID: PMC10523309 DOI: 10.3389/fmed.2023.1209886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
Objectives To assess the safety, efficacy, patients' satisfaction and acceptability of Rexon-Eye electrotherapy in treating Asian severe dry eye disease (DED) patients. Methods Prospective parallel-arm pilot study recruiting 40 DED Chinese patients with >moderate recalcitrant DED (Contact Lens Research Unit [CCLRU] > grade 2). Subjects were randomized into 2 groups, undergoing four weekly treatment sessions each: group 1 received full treatment power; group 2 received control treatment (power 1 treatment). Non-invasive tear break-up time (NIBUT), cornea fluorescein staining graded via CCLRU and Schirmer's I test were compared pre- and 2 months post-treatment. The SPEED and QUEST questionnaires that evaluated subjective symptoms and treatment satisfaction, respectively, at baseline and 2 weeks post-treatment were carried out. Tear cytokine levels in both groups were examined at 2 weeks post-treatment. Results The amount of improvement in post-treatment corneal staining in the inferior corneal zone was significant in Group 1 (p = 0.038) but not in Group 2 (p = 0.832). Group 1 eyes with worse baseline staining (total score >9.8) had a significantly greater reduction of corneal staining than those with better baseline staining (-11.7 ± 1.98 vs. -4.6 ± 2.89, p < 0.001). There were no other significant differences in NIBUT, Schirmer's 1 and cornea fluorescein staining grading within or between the groups.: Group 1 (n = 24) had improved subjective dryness scores compared to Group 2 (n = 16) (SPEED score: 6.38 + 4.16 vs. 10.0 + 6.36, p = 0.04). No significant differences were seen in 11 tear cytokine levels at 2 weeks post-treatment between the 2 groups. Conclusion In Asian DED patients treated with Rexon-Eye, inferior cornea staining showed significant improvement compared to placebo, and eyes with greater cornea staining at baseline achieved a greater improvement in staining. There were no other significant improvements in NIBUT and Schirmer's 1. Rexon-Eye also improved subjective DED scores in 41.7% of eyes without any adverse effects.
Collapse
Affiliation(s)
- Valencia Hui Xian Foo
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
- Corneal and External Eye Disease Service, Singapore National Eye Centre, Singapore, Singapore
| | - Yu-Chi Liu
- Corneal and External Eye Disease Service, Singapore National Eye Centre, Singapore, Singapore
- Eye-Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Bryan Tho
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Louis Tong
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore, Singapore
- Corneal and External Eye Disease Service, Singapore National Eye Centre, Singapore, Singapore
- Eye-Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
23
|
Han G, Lim DH, Yoo YS, Shin EH, Park JY, Kim D, Kim P, Chung TY. Transcutaneous Electrical Stimulation for the Prevention of Dry Eye Disease after Photorefractive Keratectomy: Randomized Controlled Trial. OPHTHALMOLOGY SCIENCE 2023; 3:100242. [PMID: 36685712 PMCID: PMC9853365 DOI: 10.1016/j.xops.2022.100242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/25/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Objective To evaluate the efficacy and safety of transcutaneous electrical stimulation (TES) for the prevention of dry eye after photorefractive keratectomy (PRK). Design Prospective, single-center, single-blinded, parallel group, placebo-controlled, randomized clinical trial. Participants Between February 2020 and October 2020, patients at the Samsung Medical Center scheduled to undergo PRK to correct myopia were screened and enrolled. Methods The participants in the TES group were instructed to use the electrical stimulation device (Nu Eyne 01, Nu Eyne Co) at the periocular region after the operation, whereas those in the control group were to use the sham device. Dry eye symptoms were evaluated preoperatively and postoperatively at weeks 1, 4, and 12 using the Ocular Surface Disease Index (OSDI) questionnaire, the 5-Item Dry Eye Questionnaire (DEQ-5), and the Standard Patient Evaluation for Eye Dryness II (SPEED II) questionnaire. Dry eye signs were assessed using tear break-up time (TBUT), total corneal fluorescein staining (tCFS), and total conjunctival staining score according to the National Eye Institute/Industry scale. The pain intensity was evaluated using a visual analog scale. Main Outcome Measures Primary outcomes were OSDI and TBUT. Results Twenty-four patients were enrolled and completed follow-up until the end of the study (12 patients in the TES group, 12 patients in the control group). Refractive outcomes and visual acuity were not different between the groups. No serious adverse event was reported with regard to device use. No significant difference in OSDI and SPEED II questionnaires and the DEQ-5 was observed between the groups in the 12th week after surgery. The TBUT scores 12 weeks after the surgery were 9.28 ± 6.90 seconds in the TES group and 5.98 ± 2.55 seconds in the control group with significant difference (P = 0.042). The tCFS and total conjunctival staining score were significantly lower in the TES group than in the control group at postoperative 4 weeks. Pain intensity at the first week was significantly lower in the TES group than in the control group by 65% (P = 0.011). Conclusion The application of TES is safe and effective in improving dry eye disease after PRK. Financial Disclosures The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Key Words
- Corneal nerve regeneration
- DED, dry eye disease
- DEQ-5, 5-Item Dry Eye Questionnaire
- Dry eye
- Electrostimulation
- LLT, lipid layer thickness
- NGF, nerve growth factor
- OSDI, Ocular Surface Disease Index
- PRK, photorefractive keratectomy
- Refractive surgery
- SPEED II, Standard Patient Evaluation for Eye Dryness II
- TBUT, tear break-up time
- TES, transcutaneous electrical stimulation
- UDVA, uncorrected distant visual acuity
- tCFS, total corneal fluorescein staining
Collapse
Affiliation(s)
- Gyule Han
- Department of Ophthalmology, Sungkyunkwan University, Samsung Medical Hospital, Seoul, South Korea
| | - Dong Hui Lim
- Department of Ophthalmology, Sungkyunkwan University, Samsung Medical Hospital, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Young Sik Yoo
- Department of Ophthalmology, Sungkyunkwan University, Samsung Medical Hospital, Seoul, South Korea
- Department of Ophthalmology, College of Medicine, Uijeongbu St. Mary’s Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Eun Hye Shin
- Department of Ophthalmology, Sungkyunkwan University, Samsung Medical Hospital, Seoul, South Korea
| | - Jong Yup Park
- Department of Ophthalmology, Sungkyunkwan University, Samsung Medical Hospital, Seoul, South Korea
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Dohyoung Kim
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Pyungkyu Kim
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Tae-Young Chung
- Department of Ophthalmology, Sungkyunkwan University, Samsung Medical Hospital, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
24
|
Meybodian M, Mahmoodifar S, Mandegari M, Baradaranfar M, Dadgarnia M, Vaziribozorg S. The Effect of Electrical Stimulation Therapy on Brain Derived Neurotrophic Factor in Patients with Tinnitus. Indian J Otolaryngol Head Neck Surg 2023; 75:498-501. [PMID: 37206826 PMCID: PMC10188857 DOI: 10.1007/s12070-022-03417-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/12/2022] [Indexed: 03/06/2023] Open
Abstract
Introduction: The aim of this study was to determine the effect of electric stimulation therapy on brain-derived neurotrophic factor (BDNF) in patients with tinnitus. Materials and methods: In this before-after clinical trial study, 45 patients aged 30-80 years old with tinnitus were involved. The hearing threshold, loudness, and frequency of tinnitus were assessed. Tinnitus Handicap Inventory (THI) questionnaire was completed by the patients. Before holding electrical stimulation sessions, the patients were evaluated for serum brain-derived neurotrophic factor (BDNF) level. Patients underwent five electrical stimulation sessions of 20 min for 5 consecutive days. After completion of electrical stimulation session, THI questionnaire was re-completed by the patients and they were evaluated for serum BDNF level. Results: Mean BDNF level before and after the intervention was (1238 ± 494.2) and (1148.2 ± 496.7), respectively (P = 0.04). Mean loudness score before and after the intervention was (6.36 ± 1.47) and (5.27 ± 1.68), respectively (P = 0.01). Mean THI score before and after the intervention was (58.21 ± 11.8) and (53.17 ± 15.19), respectively (p = 0.01). In patients with severe THI1, there was a significant difference between serum BDNF level (p = 0.019) and loudness (p = 0.003) before and after the intervention. However, in patients with mild, moderate, and very severe THI1, no such effect was observed (p > 0.05). Conclusion: According to the results of the present study, electrical stimulation therapy significantly decreased the mean plasma BDNF level in patients with tinnitus, especially in patients with severe tinnitus so it can be used as a marker to define the response to treatment and determine the severity of tinnitus in primary evaluations.
Collapse
Affiliation(s)
- Mojtaba Meybodian
- Department of Otolaryngology- Head and Neck Surgery, Otorhinolaryngology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Mahmoodifar
- Department of Otolaryngology- Head and Neck Surgery, Otorhinolaryngology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Mandegari
- Department of Otolaryngology- Head and Neck Surgery, Otorhinolaryngology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammadhossein Baradaranfar
- Department of Otolaryngology- Head and Neck Surgery, Otorhinolaryngology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammadhossein Dadgarnia
- Department of Otolaryngology- Head and Neck Surgery, Otorhinolaryngology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sedighe Vaziribozorg
- Department of Otolaryngology- Head and Neck Surgery, Otorhinolaryngology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
25
|
Ni L, Yao Z, Zhao Y, Zhang T, Wang J, Li S, Chen Z. Electrical stimulation therapy for peripheral nerve injury. Front Neurol 2023; 14:1081458. [PMID: 36908597 PMCID: PMC9998520 DOI: 10.3389/fneur.2023.1081458] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
Peripheral nerve injury is common and frequently occurs in extremity trauma patients. The motor and sensory impairment caused by the injury will affect patients' daily life and social work. Surgical therapeutic approaches don't assure functional recovery, which may lead to neuronal atrophy and hinder accelerated regeneration. Rehabilitation is a necessary stage for patients to recover better. A meaningful role in non-pharmacological intervention is played by rehabilitation, through individualized electrical stimulation therapy. Clinical studies have shown that electrical stimulation enhances axon growth during nerve repair and accelerates sensorimotor recovery. According to different effects and parameters, electrical stimulation can be divided into neuromuscular, transcutaneous, and functional electrical stimulation. The therapeutic mechanism of electrical stimulation may be to reduce muscle atrophy and promote muscle reinnervation by increasing the expression of structural protective proteins and neurotrophic factors. Meanwhile, it can modulate sensory feedback and reduce neuralgia by inhibiting the descending pathway. However, there are not many summary clinical application parameters of electrical stimulation, and the long-term effectiveness and safety also need to be further explored. This article aims to explore application methodologies for effective electrical stimulation in the rehabilitation of peripheral nerve injury, with simultaneous consideration for fundamental principles of electrical stimulation and the latest technology. The highlight of this paper is to identify the most appropriate stimulation parameters (frequency, intensity, duration) to achieve efficacious electrical stimulation in the rehabilitation of peripheral nerve injury.
Collapse
Affiliation(s)
- Lingmei Ni
- Infection Prevention and Control Department, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhao Yao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yifan Zhao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianfang Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jie Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Siyue Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zuobing Chen
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Dorrian RM, Berryman CF, Lauto A, Leonard AV. Electrical stimulation for the treatment of spinal cord injuries: A review of the cellular and molecular mechanisms that drive functional improvements. Front Cell Neurosci 2023; 17:1095259. [PMID: 36816852 PMCID: PMC9936196 DOI: 10.3389/fncel.2023.1095259] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that causes severe loss of motor, sensory and autonomic functions. Additionally, many individuals experience chronic neuropathic pain that is often refractory to interventions. While treatment options to improve outcomes for individuals with SCI remain limited, significant research efforts in the field of electrical stimulation have made promising advancements. Epidural electrical stimulation, peripheral nerve stimulation, and functional electrical stimulation have shown promising improvements for individuals with SCI, ranging from complete weight-bearing locomotion to the recovery of sexual function. Despite this, there is a paucity of mechanistic understanding, limiting our ability to optimize stimulation devices and parameters, or utilize combinatorial treatments to maximize efficacy. This review provides a background into SCI pathophysiology and electrical stimulation methods, before exploring cellular and molecular mechanisms suggested in the literature. We highlight several key mechanisms that contribute to functional improvements from electrical stimulation, identify gaps in current knowledge and highlight potential research avenues for future studies.
Collapse
Affiliation(s)
- Ryan M. Dorrian
- Spinal Cord Injury Research Group, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia,*Correspondence: Ryan M. Dorrian,
| | | | - Antonio Lauto
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Anna V. Leonard
- Spinal Cord Injury Research Group, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
27
|
Huynh QS, Holsinger RMD. Fiber and Electrical Field Alignment Increases BDNF Expression in SH-SY5Y Cells following Electrical Stimulation. Pharmaceuticals (Basel) 2023; 16:138. [PMID: 37259290 PMCID: PMC9960882 DOI: 10.3390/ph16020138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 09/13/2024] Open
Abstract
The limited expression of neurotrophic factors that can be included in neural tissue engineering scaffolds is insufficient for sustained neural regeneration. A localized and sustained method of introducing neurotrophic factors is required. We describe our attempt at inducing neuroblastoma cells to express trophic factors following electrical stimulation. Human SH-SY5Y neuroblastoma cells, cultured on polycaprolactone electrospun nanofibers, were electrically stimulated using a 100 mV/mm electric field. Nuclear morphology and brain-derived neurotrophic factor (BDNF) expression were analyzed. Cells were classified based on the type of fiber orientation and the alignment of these fibers in relation to the electric field. Nuclear deformation was mainly influenced by fiber orientation rather than the electrical field. Similarly, fiber orientation also induced BDNF expression. Although electrical field alone had no significant effect on BDNF expression, combining fiber orientation with electrical field resulted in BDNF expression in cells that grew on electrospun fibers that were aligned perpendicular to the electrical field.
Collapse
Affiliation(s)
- Quy-Susan Huynh
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
28
|
Juckett L, Saffari TM, Ormseth B, Senger JL, Moore AM. The Effect of Electrical Stimulation on Nerve Regeneration Following Peripheral Nerve Injury. Biomolecules 2022; 12:biom12121856. [PMID: 36551285 PMCID: PMC9775635 DOI: 10.3390/biom12121856] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Peripheral nerve injuries (PNI) are common and often result in lifelong disability. The peripheral nervous system has an inherent ability to regenerate following injury, yet complete functional recovery is rare. Despite advances in the diagnosis and repair of PNIs, many patients suffer from chronic pain, and sensory and motor dysfunction. One promising surgical adjunct is the application of intraoperative electrical stimulation (ES) to peripheral nerves. ES acts through second messenger cyclic AMP to augment the intrinsic molecular pathways of regeneration. Decades of animal studies have demonstrated that 20 Hz ES delivered post-surgically accelerates axonal outgrowth and end organ reinnervation. This work has been translated clinically in a series of randomized clinical trials, which suggest that ES can be used as an efficacious therapy to improve patient outcomes following PNIs. The aim of this review is to discuss the cellular physiology and the limitations of regeneration after peripheral nerve injuries. The proposed mechanisms of ES protocols and how they facilitate nerve regeneration depending on timing of administration are outlined. Finally, future directions of research that may provide new perspectives on the optimal delivery of ES following PNI are discussed.
Collapse
|
29
|
Yoo YS, Park S, Eun P, Park YM, Lim DH, Chung TY. Corneal Neuro-Regenerative Effect of Transcutaneous Electrical Stimulation in Rabbit Lamellar Keratectomy Model. Transl Vis Sci Technol 2022; 11:17. [PMID: 36223127 PMCID: PMC9583744 DOI: 10.1167/tvst.11.10.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study aimed to evaluate the effect of transcutaneous electrical stimulation (TES) on corneal nerve regeneration in rabbits injured from superficial lamellar keratectomy (SLK). Methods New Zealand White rabbits were used in this experimental study. To induce corneal nerve damage, SLK was performed using a 7.0-mm trephine. TES was applied for 28 days after the corneal nerve injury. Corneal sensitivity, Western blotting, real-time polymerase chain reaction (PCR), and immunofluorescence were performed to observe changes in the corneal tissue. Results In the 2-Hz and 20-Hz electrical stimulation groups, the degree of corneal wound healing increased by more than 10% compared to the control group, but no significant difference was observed. Conversely, the electrical stimulation (2-Hz or 20-Hz) group showed significantly increased corneal sensitivity compared to the control group. Western blot analysis revealed that small proline-rich protein 1A (SPRR1a), a regeneration-associated protein was significantly increased in the 2-Hz group on days 1 and 7 compared to that in the other groups. Once again, nerve regeneration in the 2-Hz group was supported by the results of PCR, in which a significant increase in the nerve growth factor (NGF) on day 1 was observed compared with the other groups. Moreover, immunofluorescence after 28 days of electrical stimulation showed significant nerve regeneration in the 2-Hz group. Conclusions TES promoted corneal nerve regeneration in rabbit SLK model. The application of electrical stimulation of 2-Hz frequency was more effective than the 20-Hz frequency, showing potential clinical applications for corneal diseases. Translational Relevance This study shows how application of TES to the eyes that exhibit corneal nerve damage can improve corneal nerve regeneration examined by histologic analysis.
Collapse
Affiliation(s)
- Young-Sik Yoo
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Ophthalmology, College of Medicine, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Sera Park
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Pyeonghwa Eun
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Young Min Park
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Dong Hui Lim
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Tae-Young Chung
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
30
|
Cheng H, Huang Y, Qian J, Meng F, Fan Y. Organic photovoltaic device enhances the neural differentiation of rat bone marrow-derived mesenchymal stem cells. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
31
|
Kim JW, Choi YY, Park SH, Ha JH, Lee HU, Kang T, Sun W, Chung BG. Microfluidic electrode array chip for electrical stimulation-mediated axonal regeneration. LAB ON A CHIP 2022; 22:2122-2130. [PMID: 35388823 DOI: 10.1039/d1lc01158h] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The precise manipulation of the neural stem cell (NSC)-derived neural differentiation is still challenging, and there is a technological barrier to regulate the axonal regeneration in a controlled manner. Here, we developed a microfluidic chip integrated with a microelectrode array as an axonal guidance platform. The microfluidic electrode array chip consisted of two compartments and a bridge microchannel that could isolate and guide the axons. We demonstrated that the NSCs were largely differentiated into neural cells as the electric field was applied to the microfluidic electrode array chip. We also confirmed the synergistic effects of the electrical stimulation (ES) and neurotrophic factor (NF) on axonal outgrowth. This microfluidic electrode array chip can serve as a central nervous system (CNS) model for axonal injury and regeneration. Therefore, it could be a potentially powerful tool for an in vitro model of the axonal regeneration.
Collapse
Affiliation(s)
- Ji Woon Kim
- Department of Biomedical Engineering, Sogang University, Seoul, Korea
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
| | - Si-Hyung Park
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea.
| | - Jang Ho Ha
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| | - Hee Uk Lee
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| | - Taewook Kang
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea.
| | - Bong Geun Chung
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| |
Collapse
|
32
|
Mutepfa AR, Hardy JG, Adams CF. Electroactive Scaffolds to Improve Neural Stem Cell Therapy for Spinal Cord Injury. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:693438. [PMID: 35274106 PMCID: PMC8902299 DOI: 10.3389/fmedt.2022.693438] [Citation(s) in RCA: 6] [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: 04/10/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) is a serious condition caused by damage to the spinal cord through trauma or disease, often with permanent debilitating effects. Globally, the prevalence of SCI is estimated between 40 to 80 cases per million people per year. Patients with SCI can experience devastating health and socioeconomic consequences from paralysis, which is a loss of motor, sensory and autonomic nerve function below the level of the injury that often accompanies SCI. SCI carries a high mortality and increased risk of premature death due to secondary complications. The health, social and economic consequences of SCI are significant, and therefore elucidation of the complex molecular processes that occur in SCI and development of novel effective treatments is critical. Despite advances in medicine for the SCI patient such as surgery and anaesthesiology, imaging, rehabilitation and drug discovery, there have been no definitive findings toward complete functional neurologic recovery. However, the advent of neural stem cell therapy and the engineering of functionalized biomaterials to facilitate cell transplantation and promote regeneration of damaged spinal cord tissue presents a potential avenue to advance SCI research. This review will explore this emerging field and identify new lines of research.
Collapse
Affiliation(s)
- Anthea R. Mutepfa
- Neural Tissue Engineering Keele, School of Life Sciences, Keele University, Keele, United Kingdom
| | - John G. Hardy
- Department of Chemistry, Lancaster University, Lancaster, United Kingdom
- Materials Science Institute, Lancaster University, Lancaster, United Kingdom
| | - Christopher F. Adams
- Neural Tissue Engineering Keele, School of Life Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
33
|
Malone IG, Kelly MN, Nosacka RL, Nash MA, Yue S, Xue W, Otto KJ, Dale EA. Closed-Loop, Cervical, Epidural Stimulation Elicits Respiratory Neuroplasticity after Spinal Cord Injury in Freely Behaving Rats. eNeuro 2022; 9:ENEURO.0426-21.2021. [PMID: 35058311 PMCID: PMC8856702 DOI: 10.1523/eneuro.0426-21.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/08/2021] [Accepted: 12/24/2021] [Indexed: 11/28/2022] Open
Abstract
Over half of all spinal cord injuries (SCIs) are cervical, which can lead to paralysis and respiratory compromise, causing significant morbidity and mortality. Effective treatments to restore breathing after severe upper cervical injury are lacking; thus, it is imperative to develop therapies to address this. Epidural stimulation has successfully restored motor function after SCI for stepping, standing, reaching, grasping, and postural control. We hypothesized that closed-loop stimulation triggered via healthy hemidiaphragm EMG activity has the potential to elicit functional neuroplasticity in spinal respiratory pathways after cervical SCI (cSCI). To test this, we delivered closed-loop, electrical, epidural stimulation (CLES) at the level of the phrenic motor nucleus (C4) for 3 d after C2 hemisection (C2HS) in freely behaving rats. A 2 × 2 Latin Square experimental design incorporated two treatments, C2HS injury and CLES therapy resulting in four groups of adult, female Sprague Dawley rats: C2HS + CLES (n = 8), C2HS (n = 6), intact + CLES (n = 6), intact (n = 6). In stimulated groups, CLES was delivered for 12-20 h/d for 3 d. After C2HS, 3 d of CLES robustly facilitated the slope of stimulus-response curves of ipsilesional spinal motor evoked potentials (sMEPs) versus nonstimulated controls. To our knowledge, this is the first demonstration of CLES eliciting respiratory neuroplasticity after C2HS in freely behaving animals. These findings suggest CLES as a promising future therapy to address respiratory deficiency associated with cSCI.
Collapse
Affiliation(s)
- Ian G Malone
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL 32611
| | - Mia N Kelly
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL 32611
- Department of Physical Therapy, University of Florida, Gainesville, FL 32611
| | - Rachel L Nosacka
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611
| | - Marissa A Nash
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611
| | - Sijia Yue
- Department of Biostatistics, University of Florida, Gainesville, FL 32611
| | - Wei Xue
- Department of Biostatistics, University of Florida, Gainesville, FL 32611
| | - Kevin J Otto
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL 32611
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL 32611
- Department of Neurology, University of Florida, Gainesville, FL 32611
- Department of Neuroscience, University of Florida, Gainesville, FL 32611
| | - Erica A Dale
- Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL 32611
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611
- McKnight Brain Institute, University of Florida, Gainesville, FL 32611
| |
Collapse
|
34
|
Vose AK, Welch JF, Nair J, Dale EA, Fox EJ, Muir GD, Trumbower RD, Mitchell GS. Therapeutic acute intermittent hypoxia: A translational roadmap for spinal cord injury and neuromuscular disease. Exp Neurol 2022; 347:113891. [PMID: 34637802 PMCID: PMC8820239 DOI: 10.1016/j.expneurol.2021.113891] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/29/2021] [Accepted: 10/03/2021] [Indexed: 01/03/2023]
Abstract
We review progress towards greater mechanistic understanding and clinical translation of a strategy to improve respiratory and non-respiratory motor function in people with neuromuscular disorders, therapeutic acute intermittent hypoxia (tAIH). In 2016 and 2020, workshops to create and update a "road map to clinical translation" were held to help guide future research and development of tAIH to restore movement in people living with chronic, incomplete spinal cord injuries. After briefly discussing the pioneering, non-targeted basic research inspiring this novel therapeutic approach, we then summarize workshop recommendations, emphasizing critical knowledge gaps, priorities for future research effort, and steps needed to accelerate progress as we evaluate the potential of tAIH for routine clinical use. Highlighted areas include: 1) greater mechanistic understanding, particularly in non-respiratory motor systems; 2) optimization of tAIH protocols to maximize benefits; 3) identification of combinatorial treatments that amplify plasticity or remove plasticity constraints, including task-specific training; 4) identification of biomarkers for individuals most/least likely to benefit from tAIH; 5) assessment of long-term tAIH safety; and 6) development of a simple, safe and effective device to administer tAIH in clinical and home settings. Finally, we update ongoing clinical trials and recent investigations of tAIH in SCI and other clinical disorders that compromise motor function, including ALS, multiple sclerosis, and stroke.
Collapse
Affiliation(s)
- Alicia K Vose
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brooks Rehabilitation, Jacksonville, FL 32216, USA
| | - Joseph F Welch
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brooks Rehabilitation, Jacksonville, FL 32216, USA
| | - Jayakrishnan Nair
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Erica A Dale
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Emily J Fox
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA; Brooks Rehabilitation, Jacksonville, FL 32216, USA
| | - Gillian D Muir
- Department of Biomedical Sciences, WCVM, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Randy D Trumbower
- Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutics Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
35
|
Yan X, Yu Y, Wang S, Xu H, He Q, Wen J, Xu J, Li K, Huang Z, Xu P. Preparation and characterization of conductive nerve guide conduit filled with dual drug-loaded nanofibers. J BIOACT COMPAT POL 2021. [DOI: 10.1177/08839115211053917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peripheral nerve injury (PNI) has become one of the common clinical diseases. How to promote the regeneration and function recovery of the damaged peripheral nerve has been the focus of attention in the medical field. Evidence suggests that the longitudinal filling of oriented fibers in nerve guide conduit (NGC) is especially beneficial to the repair of long gap PNI. In this study, polypyrrole (PPy) nanospheres (PNSs) were prepared by the soft-templating method, and mixed with poly (lactic acid) (PLA) to prepare conductive PNSs/PLA NGC, and the optimal ratio of PNSs was 4.5%. PLA and vascular endothelial growth factor (VEGF) as shell, chitosan (CS) and paeoniflorin (PF) as core, oriented coaxial nanofibers were obtained and then filled into PNSs/PLA NGC. The composite NGC has excellent mechanical properties, electrical conductivity, hydrophilic properties, and degradation properties. Besides, the successive release of VEGF and PF can play a synergistic role in promoting nerve regeneration. In vitro experiments showed that the composite NGC was nontoxic and suitable for the adhesion and proliferation of nerve cells. In addition, PNSs combined with electrical stimulation (ES) can significantly promote the differentiation and proliferation of nerve cells, which is conducive to nerve regeneration. These positive results indicate that the composite NGC is a promising candidate in the repair of long gap PNI.
Collapse
Affiliation(s)
- Xiumei Yan
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Yi Yu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Shaobing Wang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Haixing Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Qundi He
- Wuhan Mafangshan Middle School, Wuhan, China
| | - Jing Wen
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Jingyi Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Kebi Li
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Zhijun Huang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Peihu Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
36
|
Malone IG, Nosacka RL, Nash MA, Otto KJ, Dale EA. Electrical epidural stimulation of the cervical spinal cord: implications for spinal respiratory neuroplasticity after spinal cord injury. J Neurophysiol 2021; 126:607-626. [PMID: 34232771 PMCID: PMC8409953 DOI: 10.1152/jn.00625.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/07/2021] [Accepted: 06/27/2021] [Indexed: 01/15/2023] Open
Abstract
Traumatic cervical spinal cord injury (cSCI) can lead to damage of bulbospinal pathways to the respiratory motor nuclei and consequent life-threatening respiratory insufficiency due to respiratory muscle paralysis/paresis. Reports of electrical epidural stimulation (EES) of the lumbosacral spinal cord to enable locomotor function after SCI are encouraging, with some evidence of facilitating neural plasticity. Here, we detail the development and success of EES in recovering locomotor function, with consideration of stimulation parameters and safety measures to develop effective EES protocols. EES is just beginning to be applied in other motor, sensory, and autonomic systems; however, there has only been moderate success in preclinical studies aimed at improving breathing function after cSCI. Thus, we explore the rationale for applying EES to the cervical spinal cord, targeting the phrenic motor nucleus for the restoration of breathing. We also suggest cellular/molecular mechanisms by which EES may induce respiratory plasticity, including a brief examination of sex-related differences in these mechanisms. Finally, we suggest that more attention be paid to the effects of specific electrical parameters that have been used in the development of EES protocols and how that can impact the safety and efficacy for those receiving this therapy. Ultimately, we aim to inform readers about the potential benefits of EES in the phrenic motor system and encourage future studies in this area.
Collapse
Affiliation(s)
- Ian G Malone
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center (BREATHE), University of Florida, Gainesville, Florida
| | - Rachel L Nosacka
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Marissa A Nash
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Kevin J Otto
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, Florida
- Breathing Research and Therapeutics Center (BREATHE), University of Florida, Gainesville, Florida
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
- Department of Neuroscience, University of Florida, Gainesville, Florida
- Department of Neurology, University of Florida, Gainesville, Florida
- Department of Materials Science and Engineering, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Erica A Dale
- Breathing Research and Therapeutics Center (BREATHE), University of Florida, Gainesville, Florida
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
- Department of Neuroscience, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| |
Collapse
|
37
|
Application of electrical stimulation for peripheral nerve regeneration: Stimulation parameters and future horizons. INTERDISCIPLINARY NEUROSURGERY 2021. [DOI: 10.1016/j.inat.2021.101117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
38
|
Abd El Aziz AE, Sayed RH, Sallam NA, El Sayed NS. Neuroprotective Effects of Telmisartan and Nifedipine Against Cuprizone-Induced Demyelination and Behavioral Dysfunction in Mice: Roles of NF-κB and Nrf2. Inflammation 2021; 44:1629-1642. [PMID: 33709265 DOI: 10.1007/s10753-021-01447-6] [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: 12/08/2020] [Revised: 02/01/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022]
Abstract
Multiple sclerosis is a chronic inflammatory neurodegenerative disease of the central nervous system which injures the myelin sheath. Telmisartan and nifedipine are antihypertensive drugs that recently showed neuroprotective properties against neurodegenerative diseases. This study evaluated the neuroprotective effect of telmisartan or nifedipine in cuprizone-induced demyelination in mice by examining the underlying mechanisms. C57BL/6 mice received a diet containing 0.7% (w/w) cuprizone for 7 days followed by 3 weeks on a 0.2% cuprizone diet. Telmisartan (5 mg/kg/day, p.o.) or nifedipine (5 mg/kg/day, p.o.) was administered for 3 weeks starting from the second week. Telmisartan or nifedipine improved locomotor activity and enhanced motor coordination as demonstrated by open field, rotarod, and grip strength tests. Furthermore, telmisartan or nifedipine restored myelin basic protein mRNA and protein expression and increased luxol fast blue-staining intensity. Telmisartan or nifedipine attenuated cuprizone-induced oxidative stress and apoptosis by decreasing brain malondialdehyde and caspase-3 along with restoring reduced glutathione and brain-derived neurotrophic factor levels. Telmisartan or nifedipine exerted an anti-inflammatory effect by reducing the expression of nuclear factor kappa B (NF-κB p65) as well as pro-inflammatory cytokines and elevating the expression of IκB-α. In parallel, telmisartan or nifedipine upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and the levels of heme oxygenase-1 and NADPH quinone oxidoreductase 1 enzymes. In conclusion, the current study provides evidence for the protective effect of telmisartan and nifedipine in cuprizone-induced demyelination and behavioral dysfunction in mice possibly by modulating NF-κB and Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Amira E Abd El Aziz
- Center of Excellence, Arab Academy for Science and Technology and Maritime Transport, Alexandria, Egypt
| | - Rabab Hamed Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt.
| | - Nada A Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St, Cairo, 11562, Egypt
| |
Collapse
|
39
|
Fadeev FO, Bashirov FV, Markosyan VA, Izmailov AA, Povysheva TV, Sokolov ME, Kuznetsov MS, Eremeev AA, Salafutdinov II, Rizvanov AA, Lee HJ, Islamov RR. Combination of epidural electrical stimulation with ex vivo triple gene therapy for spinal cord injury: a proof of principle study. Neural Regen Res 2021; 16:550-560. [PMID: 32985487 PMCID: PMC7996027 DOI: 10.4103/1673-5374.293150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/03/2019] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Despite emerging contemporary biotechnological methods such as gene- and stem cell-based therapy, there are no clinically established therapeutic strategies for neural regeneration after spinal cord injury. Our previous studies have demonstrated that transplantation of genetically engineered human umbilical cord blood mononuclear cells producing three recombinant therapeutic molecules, including vascular endothelial growth factor (VEGF), glial cell-line derived neurotrophic factor (GDNF), and neural cell adhesion molecule (NCAM) can improve morpho-functional recovery of injured spinal cord in rats and mini-pigs. To investigate the efficacy of human umbilical cord blood mononuclear cells-mediated triple-gene therapy combined with epidural electrical stimulation in the treatment of spinal cord injury, in this study, rats with moderate spinal cord contusion injury were intrathecally infused with human umbilical cord blood mononuclear cells expressing recombinant genes VEGF165, GDNF, NCAM1 at 4 hours after spinal cord injury. Three days after injury, epidural stimulations were given simultaneously above the lesion site at C5 (to stimulate the cervical network related to forelimb functions) and below the lesion site at L2 (to activate the central pattern generators) every other day for 4 weeks. Rats subjected to the combined treatment showed a limited functional improvement of the knee joint, high preservation of muscle fiber area in tibialis anterior muscle and increased H/M ratio in gastrocnemius muscle 30 days after spinal cord injury. However, beneficial cellular outcomes such as reduced apoptosis and increased sparing of the gray and white matters, and enhanced expression of heat shock and synaptic proteins were found in rats with spinal cord injury subjected to the combined epidural electrical stimulation with gene therapy. This study presents the first proof of principle study of combination of the multisite epidural electrical stimulation with ex vivo triple gene therapy (VEGF, GDNF and NCAM) for treatment of spinal cord injury in rat models. The animal protocols were approved by the Kazan State Medical University Animal Care and Use Committee (approval No. 2.20.02.18) on February 20, 2018.
Collapse
Affiliation(s)
- Filip Olegovich Fadeev
- Department of Medical Biology and Genetics, Kazan State Medical University, Kazan, Russia
| | | | | | | | | | | | | | | | | | | | - Hyun Joon Lee
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
- Research Service, G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS, USA
| | | |
Collapse
|
40
|
Epidural Stimulation Combined with Triple Gene Therapy for Spinal Cord Injury Treatment. Int J Mol Sci 2020; 21:ijms21238896. [PMID: 33255323 PMCID: PMC7734573 DOI: 10.3390/ijms21238896] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 12/18/2022] Open
Abstract
The translation of new therapies for spinal cord injury to clinical trials can be facilitated with large animal models close in morpho-physiological scale to humans. Here, we report functional restoration and morphological reorganization after spinal contusion in pigs, following a combined treatment of locomotor training facilitated with epidural electrical stimulation (EES) and cell-mediated triple gene therapy with umbilical cord blood mononuclear cells overexpressing recombinant vascular endothelial growth factor, glial-derived neurotrophic factor, and neural cell adhesion molecule. Preliminary results obtained on a small sample of pigs 2 months after spinal contusion revealed the difference in post-traumatic spinal cord outcomes in control and treated animals. In treated pigs, motor performance was enabled by EES and the corresponding morpho-functional changes in hind limb skeletal muscles were accompanied by the reorganization of the glial cell, the reaction of stress cell, and synaptic proteins. Our data demonstrate effects of combined EES-facilitated motor training and cell-mediated triple gene therapy after spinal contusion in large animals, informing a background for further animal studies and clinical translation.
Collapse
|
41
|
Molecular and neural adaptations to neuromuscular electrical stimulation; Implications for ageing muscle. Mech Ageing Dev 2020; 193:111402. [PMID: 33189759 PMCID: PMC7816160 DOI: 10.1016/j.mad.2020.111402] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/04/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
Muscle atrophy and functional declines observed with advancing age can be minimized via various NMES protocols. Animal models have shown that NMES induces motor axon regeneration and promotes axonal outgrowth and fibre reinnervation. The activation of BDNF-trkB contributes to promotion of nerve growth and survival and mediates neuroplasticity. NMES is able to regulate muscle protein homeostasis and elevate oxidative enzyme activity.
One of the most notable effects of ageing is an accelerated decline of skeletal muscle mass and function, resulting in various undesirable outcomes such as falls, frailty, and all-cause mortality. The loss of muscle mass directly leads to functional deficits and can be explained by the combined effects of individual fibre atrophy and fibre loss. The gradual degradation of fibre atrophy is attributed to impaired muscle protein homeostasis, while muscle fibre loss is a result of denervation and motor unit (MU) remodelling. Neuromuscular electrical stimulation (NMES), a substitute for voluntary contractions, has been applied to reduce muscle mass and functional declines. However, the measurement of the effectiveness of NMES in terms of its mechanism of action on the peripheral motor nervous system and neuromuscular junction, and multiple molecular adaptations at the single fibre level is not well described. NMES mediates neuroplasticity and upregulates a number of neurotropic factors, manifested by increased axonal sprouting and newly formed neuromuscular junctions. Repeated involuntary contractions increase the activity levels of oxidative enzymes, increase fibre capillarisation and can influence fibre type conversion. Additionally, following NMES muscle protein synthesis is increased as well as functional capacity. This review will detail the neural, molecular, metabolic and functional adaptations to NMES in human and animal studies.
Collapse
|
42
|
Farokhi M, Mottaghitalab F, Saeb MR, Shojaei S, Zarrin NK, Thomas S, Ramakrishna S. Conductive Biomaterials as Substrates for Neural Stem Cells Differentiation towards Neuronal Lineage Cells. Macromol Biosci 2020; 21:e2000123. [PMID: 33015992 DOI: 10.1002/mabi.202000123] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 08/10/2020] [Indexed: 01/23/2023]
Abstract
The injuries and defects in the central nervous system are the causes of disability and death of an affected person. As of now, there are no clinically available methods to enhance neural structural regeneration and functional recovery of nerve injuries. Recently, some experimental studies claimed that the injuries in brain can be repaired by progenitor or neural stem cells located in the neurogenic sites of adult mammalian brain. Various attempts have been made to construct biomimetic physiological microenvironment for neural stem cells to control their ultimate fate. Conductive materials have been considered as one the best choices for nerve regeneration due to the capacity to mimic the microenvironment of stem cells and regulate the alignment, growth, and differentiation of neural stem cells. The review highlights the use of conductive biomaterials, e.g., polypyrrole, polyaniline, poly(3,4-ethylenedioxythiophene), multi-walled carbon nanotubes, single-wall carbon nanotubes, graphene, and graphite oxide, for controlling the neural stem cells activities in terms of proliferation and neuronal differentiation. The effects of conductive biomaterials in axon elongation and synapse formation for optimal repair of central nervous system injuries are also discussed.
Collapse
Affiliation(s)
- Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Mottaghitalab
- Nanotechnology Research CentreFaculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 14155-6451, Iran
| | | | - Shahrokh Shojaei
- Stem Cells Research CenterTissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Tehran Branch, Tehran, Iran.,Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, 1316943551, Iran
| | - Negin Khaneh Zarrin
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Sabu Thomas
- School of Chemical Sciences, MG University, Kottayam, Kerala, 686560, India
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, Department of Mechanical Engineering, National University of Singapore, Singapore, 117576, Singapore
| |
Collapse
|
43
|
Schwann Cell Role in Selectivity of Nerve Regeneration. Cells 2020; 9:cells9092131. [PMID: 32962230 PMCID: PMC7563640 DOI: 10.3390/cells9092131] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries result in the loss of the motor, sensory and autonomic functions of the denervated segments of the body. Neurons can regenerate after peripheral axotomy, but inaccuracy in reinnervation causes a permanent loss of function that impairs complete recovery. Thus, understanding how regenerating axons respond to their environment and direct their growth is essential to improve the functional outcome of patients with nerve lesions. Schwann cells (SCs) play a crucial role in the regeneration process, but little is known about their contribution to specific reinnervation. Here, we review the mechanisms by which SCs can differentially influence the regeneration of motor and sensory axons. Mature SCs express modality-specific phenotypes that have been associated with the promotion of selective regeneration. These include molecular markers, such as L2/HNK-1 carbohydrate, which is differentially expressed in motor and sensory SCs, or the neurotrophic profile after denervation, which differs remarkably between SC modalities. Other important factors include several molecules implicated in axon-SC interaction. This cell–cell communication through adhesion (e.g., polysialic acid) and inhibitory molecules (e.g., MAG) contributes to guiding growing axons to their targets. As many of these factors can be modulated, further research will allow the design of new strategies to improve functional recovery after peripheral nerve injuries.
Collapse
|
44
|
Kim HJ, Lee JS, Park JM, Lee S, Hong SJ, Park JS, Park KH. Fabrication of Nanocomposites Complexed with Gold Nanoparticles on Polyaniline and Application to Their Nerve Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:30750-30760. [PMID: 32539331 DOI: 10.1021/acsami.0c05286] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Electrically conductive materials can stimulate stem cells through electric shock and thereby contribute to the regulation of cell proliferation and differentiation. Recently, polymer-metal complexes composed of polyaniline and gold nanoparticles have emerged as novel candidates for use in regenerative medicine. By mixing two different materials, such composites maximize the benefits while alleviating the disadvantages of using either material alone. Based on their excellent conductivity, these complexes can be applied to nerve regeneration using stem cells. In this study, we investigated a method for producing hybrid nanocomposites by complexing gold nanoparticles to polyaniline and tested the resultant composites in a model of nerve regeneration. We manipulated the shape, size, and electrical conductivity of the hybrid composites by compounding the component materials at various ratios. The most efficient nanocomposite was named conductive reinforced nanocomposites (CRNc's). When the CRNc was delivered directly to cells, no cytotoxicity was observed. After the intracellular delivery of the CRNc, the stem cells were electrically stimulated using an electroporator. As a result of performing mRNA-sequencing (Seq) analysis after electrical stimulation (ES) of the CRNc-internalized cells, it was confirmed that the CRNc-internalized cells have a pattern similar to that of the positive group-induced neuron cells. In particular, microtubule-associated protein 2 is more than twice that of the control group (negative control), and the nerve fiber protein is strongly expressed as in the positive control group. In addition, we verified that neural differentiation progressed by monitoring the growth of neurites from stem cells. Together, these findings show that the CRNc can be used to induce the formation of neuron-like cells by applying ES to stem cells.
Collapse
Affiliation(s)
- Hye Jin Kim
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Jung Sun Lee
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Jong Min Park
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Sujin Lee
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Suk Jun Hong
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Ji Sun Park
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Keun-Hong Park
- Laboratory of Nano-regenerative Medical Engineering, Department of Biomedical Science, College of Life Science, CHA University, 618, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| |
Collapse
|
45
|
Wu P, Zhao Y, Chen F, Xiao A, Du Q, Dong Q, Ke M, Liang X, Zhou Q, Chen Y. Conductive Hydroxyethyl Cellulose/Soy Protein Isolate/Polyaniline Conduits for Enhancing Peripheral Nerve Regeneration via Electrical Stimulation. Front Bioeng Biotechnol 2020; 8:709. [PMID: 32719783 PMCID: PMC7347754 DOI: 10.3389/fbioe.2020.00709] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
Nerve regeneration remains a challenge to the treatment of peripheral nerve injury. Electrical stimulation (ES) is an assistant treatment to enhance recovery from peripheral nerve injury. A conductive nerve guide conduit was prepared from hydroxyethyl cellulose (HEC)/soy protein isolate (SPI)/PANI sponge (HSPS) and then the HSPS conduits were used to repair 10 mm sciatic nerve injury in rat model with or without ES, using HSPS+brain-derived neurotrophic factor (BDNF) and autografts as controls. The nerve repairing capacities were evaluated by animal experiments of behavioristics, electrophysiology, toluidine blue staining, and transmission electron microscopy (TEM) in the regenerated nerves. The results revealed that the nerve regeneration efficiency of HSPS conduits with ES (HSPS+ES) group was the best among the conduit groups but slightly lower than that of autografts group. HSPS+ES group even exhibited notably increased in the BDNF expression of regenerated nerve tissues, which was also confirmed through in vitro experiments that exogenous BDNF could promote Schwann cells proliferation and MBP protein expression. As a result, this work provided a strategy to repair nerve defect using conductive HSPS as nerve guide conduit and using ES as an extrinsic physical cue to promote the expression of endogenous BDNF.
Collapse
Affiliation(s)
- Ping Wu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yanan Zhao
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feixiang Chen
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ao Xiao
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qiaoyue Du
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qi Dong
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Meifang Ke
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao Liang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yun Chen
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Engineering Center of Natural Polymers-Based Medical Materials, Wuhan University, Wuhan, China
| |
Collapse
|
46
|
Parker J, Karantonis D, Single P. Hypothesis for the mechanism of action of ECAP-controlled closed-loop systems for spinal cord stimulation. Healthc Technol Lett 2020; 7:76-80. [PMID: 32754341 PMCID: PMC7353820 DOI: 10.1049/htl.2019.0110] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 05/06/2020] [Accepted: 05/15/2020] [Indexed: 01/14/2023] Open
Abstract
Advances in technology and improvement of efficacy for many neuromodulation applications have been achieved without understanding the relationship between the stimulation parameters and the neural activity which is generated in the nervous system. It is the neural activity that ultimately drives the therapeutic benefit and the advent of evoked compound action potential recording allows this activity to be directly measured and quantified. Closed-loop control adjusts the stimulation parameters to maintain a predetermined level of neural recruitment and has been shown to provide improved pain relief in individuals with spinal cord stimulators. However, no mechanism that relates more consistent neural recruitment to patient outcomes has been proposed. The authors propose a hypothesis that may explain the difference in efficacy between open- and closed-loop operational modes by considering the relationship between measured neural recruitment with hypothetical dose and side effect response curves. This provides a rational basis for directing clinical research and improving therapeutic systems.
Collapse
Affiliation(s)
- John Parker
- Saluda Medical Pty Ltd Artarmon, NSW, 2069, Australia.,Graduate School of Biomedical Engineering, University of New South Wales, Kensington, Australia
| | | | - Peter Single
- Saluda Medical Pty Ltd Artarmon, NSW, 2069, Australia
| |
Collapse
|
47
|
Muzzi L, Hassink G, Levers M, Jansman M, Frega M, Hofmeijer J, van Putten M, le Feber J. Mild stimulation improves neuronal survival in an in vitro model of the ischemic penumbra. J Neural Eng 2019; 17:016001. [DOI: 10.1088/1741-2552/ab51d4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
48
|
Modrak M, Talukder MAH, Gurgenashvili K, Noble M, Elfar JC. Peripheral nerve injury and myelination: Potential therapeutic strategies. J Neurosci Res 2019; 98:780-795. [PMID: 31608497 DOI: 10.1002/jnr.24538] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022]
Abstract
Traumatic peripheral nerve injury represents a major clinical and public health problem that often leads to significant functional impairment and permanent disability. Despite modern diagnostic procedures and advanced microsurgical techniques, functional recovery after peripheral nerve repair is often unsatisfactory. Therefore, there is an unmet need for new therapeutic or adjunctive strategies to promote the functional recovery in nerve injury patients. In contrast to the central nervous system, Schwann cells in the peripheral nervous system play a pivotal role in several aspects of nerve repair such as degeneration, remyelination, and axonal growth. Several non-surgical approaches, including pharmacological, electrical, cell-based, and laser therapies, have been employed to promote myelination and enhance functional recovery after peripheral nerve injury. This review will succinctly discuss the potential therapeutic strategies in the context of myelination following peripheral neurotrauma.
Collapse
Affiliation(s)
- Max Modrak
- School of Medicine & Dentistry, The University of Rochester Medical Center, Rochester, New York, USA
| | - M A Hassan Talukder
- Department of Orthopaedics & Rehabilitation, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Khatuna Gurgenashvili
- Department of Neurology, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mark Noble
- Department of Biomedical Genetics, The University of Rochester Medical Center, Rochester, New York, USA
| | - John C Elfar
- Department of Orthopaedics & Rehabilitation, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
49
|
R, Shu B, Liu X, Zhou J, Huang H, Wang J, Sun X, Qin C, An Y. Polypyrrole/polylactic acid nanofibrous scaffold cotransplanted with bone marrow stromal cells promotes the functional recovery of spinal cord injury in rats. CNS Neurosci Ther 2019; 25:951-964. [PMID: 31486601 PMCID: PMC6698972 DOI: 10.1111/cns.13135] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/17/2022] Open
Abstract
AIMS The objective of this study was to analyze the efficacy of polypyrrole/polylactic acid (PPy/PLA) nanofibrous scaffold cotransplanted with bone marrow stromal cells (BMSCs) in promoting the functional recovery in a rat spinal cord injury (SCI). METHODS Female Sprague-Dawley rats were randomly divided into three groups (n = 18/group): control group, PPy/PLA group, and PPy/PLA/BMSCs group. The SCI was induced in all rats. Consequently, rats in PPy/PLA/BMSCs group were transplanted with 1 × 105 BMSCs after implantation of PPy/PLA, while those in the PPy/PLA group were implanted with PPy/PLA only; no implantation was performed in the control group. Six weeks after surgery, immunofluorescence microscopy, electron microscope, and polymerase chain reaction (PCR) techniques were performed to assess the changes in the injured spinal cord tissues. RESULTS Electrophysiology and locomotor function testing suggested that PPy/PLA nanofibrous scaffold cotransplanted with BMSCs could promote the functional recovery of the spinal cord. Six weeks after the operation, lower amount of scar tissue was found in the PPy/PLA group compared with the control group. Abundant neurofilament (NF) and neuron-specific marker (NeuN) positive staining, and myelin formations were detected in the injured area. In addition, the transplantation of BMSCs not only improved the efficacy of PPy/PLA but also managed to survive well and was differentiated into neural and neuroglial cells. CONCLUSIONS The implantation of PPy/PLA nanofibrous scaffold and BMSCs has a great potential to restore the electrical conduction and to promote functional recovery by inhibiting the scar tissue formation, promoting axon regeneration, and bridging the gap lesion.
Collapse
Affiliation(s)
- Raynald
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine CentrePeking Union Medical College (PUMC)BeijingChina
- Department of Functional NeurosurgeryThe Third Medical Centre, Chinese PLA (People's Liberation Army) General HospitalBeijingChina
| | - Bing Shu
- Department of Neurosurgery, Beijing Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Xue‐Bin Liu
- Department of Functional NeurosurgeryThe Third Medical Centre, Chinese PLA (People's Liberation Army) General HospitalBeijingChina
| | - Jun‐Feng Zhou
- A State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and EngineeringTsinghua UniversityBeijingChina
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and EngineeringTsinghua UniversityBeijingChina
| | - Hua Huang
- Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Jing‐Yun Wang
- A State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and EngineeringTsinghua UniversityBeijingChina
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and EngineeringTsinghua UniversityBeijingChina
| | - Xiao‐Dan Sun
- A State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and EngineeringTsinghua UniversityBeijingChina
- Key Laboratory of Advanced Materials of Ministry of Education of China, School of Materials Science and EngineeringTsinghua UniversityBeijingChina
| | - Chuan Qin
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine CentrePeking Union Medical College (PUMC)BeijingChina
| | - Yi‐Hua An
- Department of Functional NeurosurgeryThe Third Medical Centre, Chinese PLA (People's Liberation Army) General HospitalBeijingChina
- Department of Neurosurgery, Beijing Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
50
|
Restoring electrical connection using a conductive biomaterial provides a new therapeutic strategy for rats with spinal cord injury. Neurosci Lett 2019; 692:33-40. [DOI: 10.1016/j.neulet.2018.10.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 11/22/2022]
|