1
|
Niu Q, Shen J, Liang W, Fan S, Yao X, Wei H, Zhang Y. Fully biodegradable ion-induced silk fibroin-based triboelectric nanogenerators with enhanced performance prevent muscle atrophy. Biomaterials 2025; 318:123185. [PMID: 39965421 DOI: 10.1016/j.biomaterials.2025.123185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/06/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Applying electrical stimulation (ES) on nerve or muscle denervation can significantly restore the nerve function and prevent muscle atrophy. The triboelectric nanogenerator (TENG) can couple the mechanical energy and electrical energy for ES. However, the triboelectric performance of fully biodegradable TENGs and the effect of ES need to be optimized and verified. Here, the triboelectric performance of silk fibroin (SF) is regulated by ions to fabricate SF-TENGs with full biodegradability, good biocompatibility, and excellent output. This SF-TENG shows a good electrostimulation recovery effect and is used for function restoration of the injured sciatic nerve and innervated muscle. Li+ effectively improves the dielectric constant and increases the positively charged ability of SF. The highest output power density of SF-TENG is 128 mW/m2, which is superior to most reported fully biodegradable TENGs. The morphology, protein expression levels, neural/muscular function are assessed to evaluate the recovery of damaged nerves and innervated muscle. The function restoration of the injured nerve and innervated muscle under ES of SF-TENG is significantly close to the normal nerve and muscle. This TENG has great potential to achieve in vivo energy generation, ES, and biodegradability as an implantable electrical stimulator for the therapy of nerve, muscle, and tissue injury.
Collapse
Affiliation(s)
- Qianqian Niu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Junjie Shen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wenhao Liang
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Suna Fan
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Xiang Yao
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Haifeng Wei
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yaopeng Zhang
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
2
|
Zhang Y, Gao Y, Zhou J, Zhang Z, Feng M, Liu Y. Advances in brain-computer interface controlled functional electrical stimulation for upper limb recovery after stroke. Brain Res Bull 2025; 226:111354. [PMID: 40280369 DOI: 10.1016/j.brainresbull.2025.111354] [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: 02/08/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Stroke often results in varying degrees of functional impairment, significantly affecting patients' quality of daily life. In recent years, brain-computer interface-controlled functional electrical stimulation has offered new therapeutic approaches for post-stroke rehabilitation. This paper reviews the application of BCI-FES in the recovery of upper limb function after stroke and explores its underlying mechanisms. By analyzing relevant studies, the aim is to provide a theoretical basis for rehabilitating upper limb function post-stroke, promote BCI-FES, and offer guidance for future clinical practice.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Dalian Medical University, College of Health-Preservation and Wellness, Dalian Medical University, China
| | - Yuling Gao
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Dalian Medical University, College of Health-Preservation and Wellness, Dalian Medical University, China
| | - Jiaqi Zhou
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Dalian Medical University, College of Health-Preservation and Wellness, Dalian Medical University, China
| | - Zhenni Zhang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Dalian Medical University, College of Health-Preservation and Wellness, Dalian Medical University, China
| | - Min Feng
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Dalian Medical University, China.
| | - Yong Liu
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Dalian Medical University, College of Health-Preservation and Wellness, Dalian Medical University, China.
| |
Collapse
|
3
|
Leckenby J, Sweitzer K, Olsen T, Mayorga-Young D, Milek D, Grobbelaar A. Current Treatments and Future Directions for Facial Paralysis. Facial Plast Surg 2025; 41:386-394. [PMID: 38955219 DOI: 10.1055/a-2358-9401] [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: 07/04/2024] Open
Abstract
Facial palsy is a condition that affects the facial nerve, the seventh of the 12 cranial nerves. Its main function is to control the muscles of facial expression. This involves the ability to express emotion through controlling the position of the mouth, the eyebrow, nostrils, and eye closure. The facial nerve also plays a key role in maintaining the posture of the mouth and as such, people with facial paralysis often have problems with drooling, speech, and dental hygiene.Due to the devastating effects on the quality of life of individuals with facial palsy, there are a multitude of various treatment options for the paralyzed face. This article reviews current management strategies and points towards promising future directions for research in the field of facial reanimation.
Collapse
Affiliation(s)
- Jonathan Leckenby
- Division of Plastic Surgery, Department of Surgery, University of Rochester, Rochester, New York, New York
- Department of Plastic and Reconstructive Surgery, Great Ormond Street Hospital for Sick Children, London, United Kingdom
| | - Keith Sweitzer
- Division of Plastic Surgery, Department of Surgery, University of Rochester, Rochester, New York, New York
| | - Timothy Olsen
- Division of Plastic Surgery, Department of Surgery, University of Rochester, Rochester, New York, New York
| | - Danielle Mayorga-Young
- Division of Plastic Surgery, Department of Surgery, University of Rochester, Rochester, New York, New York
| | - David Milek
- Division of Plastic Surgery, Department of Surgery, University of Rochester, Rochester, New York, New York
| | - Adriaan Grobbelaar
- Department of Plastic and Reconstructive Surgery, Great Ormond Street Hospital for Sick Children, London, United Kingdom
- Department for Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Sénépart O, Legay C, Hamraoui A. Managing surface energy dynamics for enhanced axonal growth: An overview of present and future challenges. BIOPHYSICS REVIEWS 2025; 6:021301. [PMID: 40321901 PMCID: PMC12045649 DOI: 10.1063/5.0237085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
To create functional neuronal circuit units during nervous system development and/or regeneration, axons are subjected to guidance signals. Expression of these signals occurs in spatiotemporal variations and is translated by the growth cone into a pathway to reach the connecting target which can be a neuron or a non-neuronal cell such as a muscle cell. This path is generated by interactions with the surrounding environment such as cells or the extracellular matrix, a complex molecular substrate. Understanding the interactions with this last component is essential to stimulate nerve regeneration in the context of motor peripheral nerve trauma, the most common source of disabilities, increasing with aging. The goal is to mimic its composition and specific characteristics using innovative biomaterials and/or implants. This review highlights some aspects of the recent findings in nerve repair. After an introduction to the peripheral nervous system, we present an overview of nerve degeneration and regeneration mechanisms before detailing the strategies used nowadays to optimize nerve (re)growth with a specific focus on the use of electric field. We discuss the advantages and limits of each option in terms of therapeutic applications.
Collapse
Affiliation(s)
| | - Claire Legay
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, F-75006 Paris, France
| | | |
Collapse
|
5
|
Tian T, Kim D, Yu K, Hartzell HC, Ward PJ. Regenerative failure of sympathetic axons contributes to deficits in functional recovery after nerve injury. Neurobiol Dis 2025; 209:106893. [PMID: 40164438 DOI: 10.1016/j.nbd.2025.106893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025] Open
Abstract
Renewed scientific interest in sympathetic modulation of muscle and neuromuscular junctions has spurred a flurry of new discoveries with major implications for motor diseases. However, the role sympathetic axons play in the persistent dysfunction that occurs after nerve injuries remains to be explored. Peripheral nerve injuries are common and lead to motor, sensory, and autonomic deficits that result in lifelong disabilities. Given the importance of sympathetic signaling in muscle metabolic health and maintaining bodily homeostasis, it is imperative to understand the regenerative capacity of sympathetic axons after injury. Therefore, we tested sympathetic axon regeneration and functional reinnervation of skin and muscle, both acute and long-term, using a battery of anatomical, pharmacological, chemogenetic, cell culture, analytical chemistry, and electrophysiological techniques. We employed several established growth-enhancing interventions, including electrical stimulation and conditioning lesion, as well as an innovative tool called bioluminescent optogenetics. Our results indicate that sympathetic regeneration is not enhanced by any of these treatments and may even be detrimental to sympathetic regeneration. Despite the complete return of motor reinnervation after sciatic nerve injury, gastrocnemius muscle atrophy and deficits in muscle cellular energy charge, as measured by relative ATP, ADP, and AMP concentrations, persisted long after injury, even with electrical stimulation. We suggest that these long-term deficits in muscle energy charge and atrophy are related to the deficiency in sympathetic axon regeneration. New studies are needed to better understand the mechanisms underlying sympathetic regeneration to develop therapeutics that can enhance the regeneration of all axon types.
Collapse
Affiliation(s)
- Tina Tian
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA 30307, USA; Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, GA 30307, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30307, USA.
| | - David Kim
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30307, USA.
| | - Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30307, USA.
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30307, USA.
| | - Patricia J Ward
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, GA 30307, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30307, USA.
| |
Collapse
|
6
|
Zhang N, Yao X, Zhang Q, Zhang C, Zheng Q, Wang Y, Shan F. Electrical stimulation promotes peripheral nerve regeneration by upregulating glycolysis and oxidative phosphorylation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167804. [PMID: 40101840 DOI: 10.1016/j.bbadis.2025.167804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/15/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025]
Abstract
Peripheral nerve injury (PNI) frequently results in motor and sensory dysfunction due to the limited regenerative capacity of axonal neurons and Schwann cells. Electrical stimulation (ES) has emerged as a promising strategy to enhance nerve regeneration; however, the underlying mechanisms, particularly those related to energy metabolism, remain poorly understood. This study aimed to investigate whether ES could promote nerve regeneration in a mouse model of PNI by modulating energy metabolism. ES was applied to the gastrocnemius and posterior thigh muscles post-sciatic nerve injury. Motor functional recovery was evaluated using gait analysis and electrophysiological test. Molecular and cellular changes in the distal nerve stumps were evaluated through Western blot and immunofluorescence staining. Nerve regeneration was assessed by neurostructural protein staining and nerve ultrastructure visualized by transmission electron microscopy. Our findings indicate that ES significantly accelerated both morphological and functional recovery following PNI. Specifically, ES upregulated energy metabolism in the sciatic nerve post-PNI by enhancing glucose uptake, glycolysis, and oxidative phosphorylation. Furthermore, ES increased the expression of neurotrophic factors and modulated the AMPK/mTOR/p70S6K signaling pathway, which are crucial for cellular metabolism and nerve regeneration. Collectively, these findings underscore the critical role of ES in modulating energy metabolism to support nerve regeneration, highlighting its potential as a clinical strategy for treating peripheral neuropathy.
Collapse
Affiliation(s)
- Nannan Zhang
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China; Department of Respiratory and Critical Care, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China
| | - Xiaoying Yao
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China
| | - Qingqing Zhang
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China
| | - Chuanji Zhang
- Shandong Daizhuang Hospital, Jining 272051, Shandong, China; Jining Key Laboratory of Neuromodulation, Jining 272051, Shandong, China
| | - Qian Zheng
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China
| | - Yuzhong Wang
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China; Department of Neurology, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China.
| | - Fangzhen Shan
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining 272029, Shandong, China.
| |
Collapse
|
7
|
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
|
8
|
Stephens T, Bristol S, Chapman KM, Doherty C, Seal A, Krauss EM, Cunningham C, O'Connor R, Jack K, Berger MJ. Understanding surgical decision-making in patients with traumatic upper extremity peripheral nerve injury: A retrospective cohort study. J Plast Reconstr Aesthet Surg 2025; 104:407-413. [PMID: 40174258 DOI: 10.1016/j.bjps.2025.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE Careful patient selection and optimal surgical timing are essential to the success of nerve transfers. It is important to understand what factors contribute to this decision-making. The purpose of this study was to describe the characteristics of patients referred to interdisciplinary peripheral nerve clinics with traumatic upper extremity injuries and compare those who went on to nerve transfer surgery with those who did not. METHODS Patient and injury characteristics, preoperative physical examination and electrodiagnostic findings, and patient-reported outcome measures were examined. Inclusion criteria were subjects ≥18 years of age presenting to an interdisciplinary peripheral nerve clinic with traumatic upper extremity peripheral nerve injuries. Subjects were stratified into surgical and non-surgical groups for comparison. RESULTS Eighty-three subjects met the inclusion criteria, and 36 subjects received nerve transfer surgery. More male subjects went on to have surgery than female subjects. The surgical group demonstrated a significantly higher ratio of weak and denervated muscle groups than the non-surgical group (p < 0.05). No other statistically significant differences were identified between operative and non-operatively managed subjects. CONCLUSION Subjects that received nerve transfer surgery demonstrated a significantly higher ratio of weak and denervated muscles than those managed non-surgically, and males were disproportionately represented in the surgical group. These findings suggest that anticipated motor recovery is the most important factor driving surgical decision-making and that male subjects may be more likely to proceed with surgery. Understanding which patients undergo nerve transfer surgery allows clinicians to interrogate their decision-making, address patient-related barriers to surgery, and better understand surgical outcomes.
Collapse
Affiliation(s)
- Trina Stephens
- Division of Plastic Surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sean Bristol
- Division of Plastic Surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada
| | - Kristine M Chapman
- British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada; Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher Doherty
- Division of Plastic Surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada
| | - Alexander Seal
- Division of Plastic Surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada
| | - Emily M Krauss
- Division of Plastic Surgery, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Cameron Cunningham
- British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada; Division of Physical Medicine & Rehabilitation, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Russell O'Connor
- British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada; ICORD-Blusson Spinal Cord Centre, Vancouver, British Columbia, Canada
| | - Kristin Jack
- British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada; Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael J Berger
- British Columbia Centre for Complex Nerve Injury, Vancouver, British Columbia, Canada; Division of Physical Medicine & Rehabilitation, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
9
|
Lv S, Wu Z, Huang Y, Wu P, Shao J, Wu J, Zhong K, Zhou L, Wu W. Chronic motoneuronal activation enhanced axonal regeneration and functional recovery after brachial plexus injury. J Orthop Translat 2025; 52:40-54. [PMID: 40235635 PMCID: PMC11999476 DOI: 10.1016/j.jot.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 04/17/2025] Open
Abstract
Background Brachial plexus injury (BPI) leads to significant impairment of upper limb motor function, primarily due to progressive atrophy of denervated muscles resulting from the slow rate of axonal regeneration. Therefore, identifying strategies to accelerate axon extension is of critical importance. Methods In this study, we first established a mouse model of brachial plexus injury and employed chemogenetic approaches to specifically activate C6 spinal motoneurons. We then assessed axonal regeneration and motor function recovery in the injured mice through behavioral tests, morphological analyses, and electrophysiological detection. Results We found that the AAV9-hM3Dq virus efficiently transduced motoneurons, and CNO administration robustly activated mature hM3Dq+ motoneurons in vivo. Chronic chemogenetic activation significantly enhanced the regeneration of spinal motoneurons injured by ventral root crush, accelerated axon extension, and improved axonal remyelination, resulting in increased axon size. This activation also facilitated the formation of new neuromuscular junctions (NMJs) in adult motoneurons and reduced muscle atrophy. Furthermore, it promoted electrophysiological recovery of the motor unit and improved overall motor function. Conclusion Chemogenetic activation of adult motoneurons can robustly enhances axon growth and mediate better behavioral recovery. These findings highlight the therapeutic potential of chemogenetic neuronal activation in promoting functional recovery following nerve injury. The translational potential of this article We have established a chronic chemogenetic method to activate hM3Dq+ motor neurons after brachial plexus injury, which accelerates axonal regeneration and enhances functional recovery. This strategy holds promise as a clinical therapeutic approach for treating nervous system injuries.
Collapse
Affiliation(s)
- Shiqin Lv
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Zizhuo Wu
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Yu Huang
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Pingzhen Wu
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Jianqing Shao
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Jiajia Wu
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Ke Zhong
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangdong, 510102, China, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou, Guangdong, 510102, China
| | - Lihua Zhou
- Department of Anatomy, School of Medicine (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, 518107, China, No.66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Wutian Wu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education Joint International Research Laboratory of Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong, 510632, China, No.601, West Huangpu Avenue, Tianhe District, Guangzhou, Guangdong, 510632, China
- Jiangsu RE-STEM Biotechnology Co., Ltd., Building A1, Yuewang Zhihuigu, 1463 Wuzhong Avenue, Suzhou, Jiangsu, 215104, China
| |
Collapse
|
10
|
Grosu-Bularda A, Vancea CV, Hodea FV, Cretu A, Bordeanu-Diaconescu EM, Dumitru CS, Ratoiu VA, Teodoreanu RN, Lascar I, Hariga CS. Optimizing Peripheral Nerve Regeneration: Surgical Techniques, Biomolecular and Regenerative Strategies-A Narrative Review. Int J Mol Sci 2025; 26:3895. [PMID: 40332790 PMCID: PMC12027958 DOI: 10.3390/ijms26083895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Peripheral nerve injury disrupts the function of the peripheral nervous system, leading to sensory, motor, and autonomic deficits. While peripheral nerves possess an intrinsic regenerative capacity, complete sensory and motor recovery remains challenging due to the unpredictable nature of the healing process, which is influenced by the extent of the injury, age, and timely intervention. Recent advances in microsurgical techniques, imaging technologies, and a deeper understanding of nerve microanatomy have enhanced functional outcomes in nerve repair. Nerve injury initiates complex pathophysiological responses, including Wallerian degeneration, macrophage activation, Schwann cell dedifferentiation, and axonal sprouting. Complete nerve disruptions require surgical intervention to restore nerve continuity and function. Direct nerve repair is the gold standard for clean transections with minimal nerve gaps. However, in cases with larger nerve gaps or when direct repair is not feasible, alternatives such as autologous nerve grafting, vascularized nerve grafts, nerve conduits, allografts, and nerve transfers may be employed. Autologous nerve grafts provide excellent biocompatibility but are limited by donor site morbidity and availability. Vascularized grafts are used for large nerve gaps and poorly vascularized recipient beds, while nerve conduits serve as a promising solution for smaller gaps. Nerve transfers are utilized when neither direct repair nor grafting is possible, often involving re-routing intact regional nerves to restore function. Nerve conduits play a pivotal role in nerve regeneration by bridging nerve gaps, with significant advancements made in material composition and design. Emerging trends in nerve regeneration include the use of 3D bioprinting for personalized conduits, gene therapy for targeted growth factor delivery, and nanotechnology for nanofiber-based conduits and stem cell therapy. Advancements in molecular sciences have provided critical insights into the cellular and biochemical mechanisms underlying nerve repair, leading to targeted therapies that enhance axonal regeneration, remyelination, and functional recovery in peripheral nerve injuries. This review explores the current strategies for the therapeutic management of peripheral nerve injuries, highlighting their indications, benefits, and limitations, while emphasizing the need for tailored approaches based on injury severity and patient factors.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Vladimir Vancea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Andrei Cretu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Eliza-Maria Bordeanu-Diaconescu
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Catalina-Stefania Dumitru
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Vladut-Alin Ratoiu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Razvan-Nicolae Teodoreanu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Ioan Lascar
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
11
|
Wang H, Guo J, Zhang Y, Fu Z, Yao Y. Closed-loop rehabilitation of upper-limb dyskinesia after stroke: from natural motion to neuronal microfluidics. J Neuroeng Rehabil 2025; 22:87. [PMID: 40253334 PMCID: PMC12008995 DOI: 10.1186/s12984-025-01617-9] [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: 12/04/2024] [Accepted: 03/27/2025] [Indexed: 04/21/2025] Open
Abstract
This review proposes an innovative closed-loop rehabilitation strategy that integrates multiple subdomains of stroke science to address the global challenge of upper-limb dyskinesia post-stroke. Despite advancements in neural remodeling and rehabilitation research, the compartmentalization of subdomains has limited the effectiveness of current rehabilitation strategies. Our approach unites key areas-including the post-stroke brain, upper-limb rehabilitation robotics, motion sensing, metrics, neural microfluidics, and neuroelectronics-into a cohesive framework designed to enhance upper-limb motion rehabilitation outcomes. By leveraging cutting-edge technologies such as lightweight rehabilitation robotics, advanced motion sensing, and neural microfluidic models, this strategy enables real-time monitoring, adaptive interventions, and personalized rehabilitation plans. Furthermore, we explore the potential of closed-loop systems to drive neural plasticity and functional recovery, offering a transformative perspective on stroke rehabilitation. Finally, we discuss future directions, emphasizing the integration of emerging technologies and interdisciplinary collaboration to advance the field. This review highlights the promise of closed-loop strategies in achieving unprecedented integration of subdomains and improving post-stroke upper-limb rehabilitation outcomes.
Collapse
Affiliation(s)
- Honggang Wang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Junlong Guo
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Yangqi Zhang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China
| | - Ze Fu
- Institute of Biological and Medical Technology, Harbin Institute of Technology (Weihai), Weihai, 264200, China
| | - Yufeng Yao
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
12
|
Burrell JC, Ali ZS, Zager EL, Rosen JM, Tatarchuk MM, Cullen DK. Engineering the Future of Restorative Clinical Peripheral Nerve Surgery. Adv Healthc Mater 2025:e2404293. [PMID: 40166822 DOI: 10.1002/adhm.202404293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/25/2025] [Indexed: 04/02/2025]
Abstract
Peripheral nerve injury is a significant clinical challenge, often leading to permanent functional deficits. Standard interventions, such as autologous nerve grafts or distal nerve transfers, require sacrificing healthy nerve tissue and typically result in limited motor or sensory recovery. Nerve regeneration is complex and influenced by several factors: 1) the regenerative capacity of proximal neurons, 2) the ability of axons and support cells to bridge the injury, 3) the capacity of Schwann cells to maintain a supportive environment, and 4) the readiness of target muscles or sensory organs for reinnervation. Emerging bioengineering solutions, including biomaterials, drug delivery systems, fusogens, electrical stimulation devices, and tissue-engineered products, aim to address these challenges. Effective translation of these therapies requires a deep understanding of the physiology and pathology of nerve injury. This article proposes a comprehensive framework for developing restorative strategies that address all four major physiological responses in nerve repair. By implementing this framework, we envision a paradigm shift that could potentially enable full functional recovery for patients, where current approaches offer minimal hope.
Collapse
Affiliation(s)
- Justin C Burrell
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration & Restoration, CMC VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Oral and Maxillofacial Surgery & Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Zarina S Ali
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Nerve Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eric L Zager
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Nerve Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Joseph M Rosen
- Division of Plastic Surgery, Dartmouth-Hitchcock Medical Center, Dartmouth College, Lebanon, NH, 03766, USA
| | - Mykhailo M Tatarchuk
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration & Restoration, CMC VA Medical Center, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration & Restoration, CMC VA Medical Center, Philadelphia, PA, 19104, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Axonova Medical, LLC, Philadelphia, PA, 19104, USA
| |
Collapse
|
13
|
Koopman JE, de Groot LG, Zuidam JM, Duraku LS, Hooijmans CR, Hundepool CA. Does short-term intraoperative electrical stimulation enhance nerve regeneration following peripheral nerve repair? A systematic review and meta-analysis. J Plast Reconstr Aesthet Surg 2025:S1748-6815(25)00219-0. [PMID: 40199698 DOI: 10.1016/j.bjps.2025.03.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Although intraoperative electrical nerve stimulation appears to be a promising neuroenhancing adjunct to peripheral nerve repair, insight into its effects on nerve regeneration is essential to advocate its application in clinical settings. OBJECTIVE This study examined whether electrical stimulation during microsurgical repair of peripheral nerve injury results in enhanced nerve regeneration compared to suture repair alone in experimental animals. METHODS A systematic search in Embase, MEDLINE, Web of Science, and Google Scholar databases was performed from inception to March 22, 2024. The search included animal studies assessing outcomes following peripheral nerve repair with and without intraoperative electrical stimulation. Outcomes were subdivided into 4 categories: motor function, sensory function, electrophysiology, and histology. We calculated standardized mean differences and combined these using random effects models to estimate the overall effect. The risk of bias was assessed using the SYRCLE tool. RESULTS From 3615 references, 21 articles were included. Thirteen studies evaluated motor functional outcomes and showed that electrical stimulation improved functional index, muscle mass, muscle force, footstep accuracy, footprint, and joint angle measures. Six studies examined sensory function and found that electrical stimulation improved mechanical algesimetry. Nine studies assessed electrophysiology outcomes. Although conduction velocity did not differ between the groups, electrical stimulation resulted in a higher amplitude and lower latency. Twenty studies evaluated the histological outcomes and demonstrated increased axon count and myelin thickness, whereas axon diameter and G-ratio did not differ. DISCUSSION The results suggest that intraoperative electrical stimulation following peripheral nerve repair accelerates and improves nerve regeneration compared with nerve repair alone. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023455066.
Collapse
Affiliation(s)
- Jaimy E Koopman
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Lucas G de Groot
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jelle M Zuidam
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liron S Duraku
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Caroline A Hundepool
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
14
|
Lopes Alves R, Zortea M, Mayor D, Watson T, Steffert T. Effect of Different Frequencies of Transcutaneous Electrical Acupoint Stimulation (TEAS) on EEG Source Localization in Healthy Volunteers: A Semi-Randomized, Placebo-Controlled, Crossover Study. Brain Sci 2025; 15:270. [PMID: 40149791 PMCID: PMC11940437 DOI: 10.3390/brainsci15030270] [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: 12/14/2024] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Transcutaneous electrical acupoint stimulation (TEAS), also known as transcutaneous electroacupuncture stimulation, delivers electrical pulses to the skin over acupuncture points ("acupoints") via surface electrodes. Electroencephalography (EEG) is an important tool for assessing the changes in the central nervous system (CNS) that may result from applying different TEAS frequencies peripherally-i.e., acting via the peripheral nervous system (PNS)-and determining how these influence cerebral activity and neural plasticity. Methods: A total of 48 healthy volunteers were allocated in a semi-randomized crossover study to receive four different TEAS frequencies: 2.5 pulses per second (pps); 10 pps; 80 pps; and sham (160 pps at a low, clinically ineffective amplitude). TEAS was applied for 20 min to each hand at the acupuncture point Hegu (LI4). The EEG was recorded during an initial 5 min baseline recording, then during TEAS application, and after stimulation for a further 15 min, separated into three periods of 5 min (initial, intermediate, and final) in order to assess post-stimulation changes. Source localization analysis was conducted for the traditional five EEG frequency bands: delta (0.1-3.9 Hz), theta (4-7.9 Hz), alpha (8-13 Hz), beta (14-30 Hz), and gamma (30.1-45 Hz). Results: Within-group source localization analyses of EEG data showed that during the initial 5 min post-stimulation, theta oscillations in the 2.5 pps TEAS group increased over the parahippocampal gyrus (t = 4.42, p < 0.01). The 10 pps TEAS group exhibited decreased alpha rhythms over the inferior parietal gyrus (t = -4.20, p < 0.05), whereas the sham (160 pps) TEAS group showed decreased delta rhythms over the postcentral gyrus (t = -3.97, p < 0.05). During the intermediate 5 min post-stimulation, the increased theta activity over the left parahippocampal gyrus (BA27) remained in the 2.5 pps TEAS group (t = 3.97, p < 0.05). However, diminished alpha rhythms were observed in the 10 pps TEAS group over the postcentral gyrus (t = -4.20, p < 0.01), as well as in the delta rhythms in the sham (160 pps) TEAS group in the same area (t = -4.35, p < 0.01). In the final 5 min post-stimulation, reduced alpha rhythms were exhibited over the insula in the 10 pps TEAS group (t = -4.07, p < 0.05). Interaction effects of condition by group demonstrate decreased alpha rhythms in the 10 pps TEAS group over the supramarginal gyrus during the initial 5 min post-stimulation (t = -4.31, p < 0.05), and decreased delta rhythms over the insula in the sham TEAS group during the final 5 min post-stimulation (t = -4.42, p < 0.01). Conclusions: This study revealed that low TEAS frequencies of 2.5 pps and 10 pps modulate theta and alpha oscillations over the brain areas related to emotional and attentional processes driven by external stimuli, as well as neural synchronization of delta rhythms in the sham group in brain areas related to stimulus expectation at baseline. It is hoped that these findings will stimulate further research in order to evaluate such TEAS modulation effects in clinical patients.
Collapse
Affiliation(s)
| | | | - David Mayor
- School of Health and Social Work, University of Hertfordshire, Hatfield AL10 9AB, UK; (D.M.); (T.W.)
| | - Tim Watson
- School of Health and Social Work, University of Hertfordshire, Hatfield AL10 9AB, UK; (D.M.); (T.W.)
| | - Tony Steffert
- MindSpire, Napier House, 14-16 Mount Ephraim Rd., Tunbridge Wells TN1 1EE, UK
- School of Life, Health and Chemical Sciences, Walton Hall, The Open University, Milton Keynes MK7 6AA, UK
| |
Collapse
|
15
|
Linh VTN, Han S, Koh E, Kim S, Jung HS, Koo J. Advances in wearable electronics for monitoring human organs: Bridging external and internal health assessments. Biomaterials 2025; 314:122865. [PMID: 39357153 DOI: 10.1016/j.biomaterials.2024.122865] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/06/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Devices used for diagnosing disease are often large, expensive, and require operation by trained professionals, which can result in delayed diagnosis and missed opportunities for timely treatment. However, wearable devices are being recognized as a new approach to overcoming these difficulties, as they are small, affordable, and easy to use. Recent advancements in wearable technology have made monitoring information possible from the surface of organs like the skin and eyes, enabling accurate diagnosis of the user's internal status. In this review, we categorize the body's organs into external (e.g., eyes, oral cavity, neck, and skin) and internal (e.g., heart, brain, lung, stomach, and bladder) organ systems and introduce recent developments in the materials and designs of wearable electronics, including electrochemical and electrophysiological sensors applied to each organ system. Further, we explore recent innovations in wearable electronics for monitoring of deep internal organs, such as the heart, brain, and nervous system, using ultrasound, electrical impedance tomography, and temporal interference stimulation. The review also addresses the current challenges in wearable technology and explores future directions to enhance the effectiveness and applicability of these devices in medical diagnostics. This paper establishes a framework for correlating the design and functionality of wearable electronics with the physiological characteristics and requirements of various organ systems.
Collapse
Affiliation(s)
- Vo Thi Nhat Linh
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea
| | - Seunghun Han
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul, 02841, South Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Eunhye Koh
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea
| | - Sumin Kim
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul, 02841, South Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea
| | - Ho Sang Jung
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon, 51508, South Korea; Advanced Materials Engineering, University of Science and Technology (UST), Daejeon, 34113, South Korea; School of Convergence Science and Technology, Medical Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea.
| | - Jahyun Koo
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul, 02841, South Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
16
|
Zapadka TE, Tran NM, Demb JB. Optic nerve injury impairs intrinsic mechanisms underlying electrical activity in a resilient retinal ganglion cell. J Physiol 2025. [PMID: 39985791 DOI: 10.1113/jp286414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 01/22/2025] [Indexed: 02/24/2025] Open
Abstract
Retinal ganglion cells (RGCs) are the sole output neurons of the retina and convey visual information to the brain via their axons in the optic nerve. Following injury to the optic nerve, RGC axons degenerate and many cells die. For example, a model of axon injury, the optic nerve crush (ONC), kills ∼80% of RGCs after 2 weeks. Surviving cells are biased towards 'resilient' types, including several with sustained firing to light stimulation. RGC survival may depend on activity, and there is limited understanding of how or why activity changes following optic nerve injury. Here we quantified the electrophysiological properties of a highly resilient RGC type, the sustained ON-Alpha (AlphaONS) RGC, 7 days after ONC with extracellular and whole-cell patch clamp recording. Both light- and current-driven firing were reduced after ONC, but synaptic inputs were largely intact. Resting membrane potential and input resistance were relatively unchanged, while voltage-gated currents were impaired, including a reduction in voltage-gated sodium channel current and channel density in the axon initial segment. Hyperpolarization or chelation of intracellular calcium partially rescued firing rates. Extracellular recordings at 3 days following ONC showed normal light-evoked firing from AlphaONS RGCs and other Alpha RGCs, including susceptible types. These data suggest that an injured resilient RGC reduces its activity by 1 week after injury as a consequence of reduced voltage-gated current and downregulation of intrinsic excitability via a Ca2+-dependent mechanism. Reduced excitability may be due to degradation of the axon but could also be energetically beneficial, preserving energy for survival and regeneration. KEY POINTS: Retinal ganglion cell (RGC) types show diverse rates of survival after axon injury. A resilient RGC type (sustained ON-Alpha RGC) maintains its synaptic inputs 1 week after injury. The resilient RGC type shows diminished firing and reduced expression of axon initial segment genes 1 week after injury Activity deficits reflect dysfunction of intrinsic properties (Na+ channels, intracellular Ca2+), not changes to synaptic input. Both resilient and susceptible Alpha RGC types show intact firing at 3 days after injury, suggesting that activity at this time point does not predict resilience.
Collapse
Affiliation(s)
- Thomas E Zapadka
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Nicholas M Tran
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan B Demb
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
- Department of Ophthalmology and Visual Science, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
17
|
Candan B, Gungor S. Current and Evolving Concepts in the Management of Complex Regional Pain Syndrome: A Narrative Review. Diagnostics (Basel) 2025; 15:353. [PMID: 39941283 PMCID: PMC11817358 DOI: 10.3390/diagnostics15030353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Complex regional pain syndrome (CRPS) is characterized by severe pain and reduced functionality, which can significantly affect an individual's quality of life. The current treatment of CRPS is challenging. However, recent advances in diagnostic and treatment methods show promise for improving patient outcomes. This review aims to place the question of CRPS in a broader context and highlight the objectives of the research for future directions in the management of CRPS. Methods: This study involved a comprehensive literature review. Results: Research has identified three primary pathophysiological pathways that may explain the clinical variability observed in CRPS: inflammatory mechanisms, vasomotor dysfunction, and maladaptive neuroplasticity. Investigations into these pathways have spurred the development of novel diagnostic and treatment strategies focused on N-Methyl-D-aspartate Receptor Antagonists (NMDA), Toll-like receptor 4 (TLR-4), α1 and α2 adrenoreceptors, as well as the identification of microRNA (miRNA) biomarkers. Treatment methods being explored include immune and glial-modulating agents, intravenous immunoglobulin (IVIG) therapy, plasma exchange therapy, and neuromodulation techniques. Additionally, there is ongoing debate regarding the efficacy of other treatments, such as free radical scavengers, alpha-lipoic acid (ALA), dimethyl fumarate (DMF), adenosine monophosphate-activated protein kinase (AMPK) activators such as metformin, and phosphodiesterase-5 inhibitors such as tadalafil. Conclusions: The controversies surrounding the mechanisms, diagnosis, and treatment of CRPS have prompted researchers to investigate new approaches aimed at enhancing understanding and management of the condition, with the goal of alleviating symptoms and reducing associated disabilities.
Collapse
Affiliation(s)
- Burcu Candan
- Department of Anesthesiology and Reanimation, Bahçeşehir University Göztepe Medical Park Hospital, 34732 Istanbul, Türkiye
| | - Semih Gungor
- Division of Musculoskeletal and Interventional Pain Management, Department of Anesthesiology, Critical Care and Pain Management, Hospital for Special Surgery, New York, NY 10021, USA;
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
18
|
Li Z, Gao Y, Chen X, Xu L, Li Z, Chai R. Study on Recovery Strategy of Hearing Loss & SGN Regeneration Under Physical Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410919. [PMID: 39716878 PMCID: PMC11791950 DOI: 10.1002/advs.202410919] [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: 09/09/2024] [Revised: 11/18/2024] [Indexed: 12/25/2024]
Abstract
The World Health Organization (WHO) reports that by 2050, nearly 2.5 billion people are expected to have some degree of hearing loss (HL) and at least 700 million will need hearing rehabilitation. Therefore, there is an urgent need to develop treatment strategies for HL. At present, the main treatment strategies for HL are hearing aids and cochlear implants (CIs), which cannot achieve a radical cure for HL. Relevant studies have shown that the most fundamental treatment strategy for sensorineural hearing loss (SNHL) is to regenerate hair cells and spiral ganglion neurons (SGNs) through stem cells to repair the structure and function of cochlea. In addition, physical stimulation strategies, such as electricity, light, and magnetism have also been used to promote SGN regeneration. This review systematically introduces the classification, principle and latest progress of the existing hearing treatment strategies and summarizes the advantages and disadvantages of each strategy. The research progress of physical regulation mechanism is discussed in detail. Finally, the problems in HL repair strategies are summarized and the future development direction is prospected, which could provide new ideas and technologies for the optimization of hearing treatment strategies and the research of SGN repair and regeneration through physical regulation.
Collapse
Affiliation(s)
- Zhe Li
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
| | - Yijia Gao
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
| | - Xingyu Chen
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
| | - Lei Xu
- Department of Otolaryngology‐Head and Neck SurgeryShandong Provincial ENT HospitalShandong UniversityJinan250022China
| | - Zhou Li
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
- School of Nanoscience and EngineeringUniversity of Chinese Academy of SciencesBeijing100049China
| | - Renjie Chai
- Department of NeurologyAerospace Center HospitalSchool of LifeBeijing Institute of TechnologyBeijing100081China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologySchool of MedicineAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
| |
Collapse
|
19
|
Borah R, Diez Clarke D, Upadhyay J, Monaghan MG. From innovation to clinic: Emerging strategies harnessing electrically conductive polymers to enhance electrically stimulated peripheral nerve repair. Mater Today Bio 2025; 30:101415. [PMID: 39816667 PMCID: PMC11733191 DOI: 10.1016/j.mtbio.2024.101415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
Peripheral nerve repair (PNR) is a major healthcare challenge due to the limited regenerative capacity of the nervous system, often leading to severe functional impairments. While nerve autografts are the gold standard, their implications are constrained by issues such as donor site morbidity and limited availability, necessitating innovative alternatives like nerve guidance conduits (NGCs). However, the inherently slow nerve growth rate (∼1 mm/day) and prolonged neuroinflammation, delay recovery even with the use of passive (no-conductive) NGCs, resulting in muscle atrophy and loss of locomotor function. Electrical stimulation (ES) has the ability to enhance nerve regeneration rate by modulating the innate bioelectrical microenvironment of nerve tissue while simultaneously fostering a reparative environment through immunoregulation. In this context, electrically conductive polymer (ECP)-based biomaterials offer unique advantages for nerve repair combining their flexibility, akin to traditional plastics, and mixed ionic-electronic conductivity, similar to ionically conductive nerve tissue, as well as their biocompatibility and ease of fabrication. This review focuses on the progress, challenges, and emerging techniques for integrating ECP based NGCs with ES for functional nerve regeneration. It critically evaluates the various approaches using ECP based scaffolds, identifying gaps that have hindered clinical translation. Key challenges discussed include designing effective 3D NGCs with high electroactivity, optimizing ES modules, and better understanding of immunoregulation during nerve repair. The review also explores innovative strategies in material development and wireless, self-powered ES methods. Furthermore, it emphasizes the need for non-invasive ES delivery methods combined with hybrid ECP based neural scaffolds, highlighting future directions for advancing preclinical and clinical translation. Together, ECP based NGCs combined with ES represent a promising avenue for advancing PNR and improving patient outcomes.
Collapse
Affiliation(s)
- Rajiv Borah
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel Diez Clarke
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - Jnanendra Upadhyay
- Department of Physics, Dakshin Kamrup College, Kamrup, Assam, 781125, India
| | - Michael G. Monaghan
- Discipline of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland
- CÚRAM, Research Ireland Centre for Research in Medical Devices, University of Galway, H91 W2TY Galway, Ireland
| |
Collapse
|
20
|
Tian T, Patel K, Kim D, SiMa H, Harris AR, Owyoung JN, Ward PJ. Conditioning electrical stimulation fails to enhance sympathetic axon regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.02.03.527071. [PMID: 36778305 PMCID: PMC9915730 DOI: 10.1101/2023.02.03.527071] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Peripheral nerve injuries are common, and there is a critical need for the development of novel treatments to complement surgical repair. Conditioning electrical stimulation (CES) is a novel variation of the well-studied perioperative electrical stimulation treatment paradigm. CES is a clinically attractive alternative because of its ability to be performed at the bedside prior to a scheduled nerve repair surgery. Although 60 minutes of CES has been shown to enhance motor and sensory axon regeneration, the effects of CES on sympathetic regeneration are unknown. We investigated how two clinically relevant CES paradigms (10 minutes and 60 minutes) impact sympathetic axon regeneration and distal target reinnervation. Our results indicate that the growth of sympathetic axons is inhibited by CES at acute time points, and at a longer survival time point post-injury, there is no difference between sham CES and the CES groups. We conclude sympathetic axons may retain some regenerative ability, but no enhancement is exhibited after CES, which may be accounted for by the inability of the electrical stimulation paradigm to recruit the small-caliber sympathetic axons into activity. Furthermore, 10-minute CES did not enhance motor and sensory regeneration with a direct repair, and neither 60-minute nor 10-minute CES enhanced motor and sensory regeneration through a graft. Further studies will be needed to optimize electrical stimulation parameters to enhance the regeneration of all neuron types.
Collapse
|
21
|
Tian T, Kim D, Yu K, Hartzell HC, Ward PJ. Regenerative failure of sympathetic axons contributes to deficits in functional recovery after nerve injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631956. [PMID: 39829867 PMCID: PMC11741411 DOI: 10.1101/2025.01.08.631956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Renewed scientific interest in sympathetic modulation of muscle and neuromuscular junctions has spurred a flurry of new discoveries with major implications for motor diseases. However, the role sympathetic axons play in the persistent dysfunction that occurs after nerve injuries remains to be explored. Peripheral nerve injuries are common and lead to motor, sensory, and autonomic deficits that result in lifelong disabilities. Given the importance of sympathetic signaling in muscle metabolic health and maintaining bodily homeostasis, it is imperative to understand the regenerative capacity of sympathetic axons after injury. Therefore, we tested sympathetic axon regeneration and functional reinnervation of skin and muscle, both acute and long-term, using a battery of anatomical, pharmacological, chemogenetic, cell culture, analytical chemistry, and electrophysiological techniques. We employed several established growth-enhancing interventions, including electrical stimulation and conditioning lesion, as well as an innovative tool called bioluminescent optogenetics. Our results indicate that sympathetic regeneration is not enhanced by any of these treatments and may even be detrimental to sympathetic regeneration. Despite the complete return of motor reinnervation after sciatic nerve injury, gastrocnemius muscle atrophy and deficits in muscle cellular energy charge, as measured by relative ATP, ADP, and AMP concentrations, persisted long after injury, even with electrical stimulation. We suggest that these long-term deficits in muscle energy charge and atrophy are related to the deficiency in sympathetic axon regeneration. New studies are needed to better understand the mechanisms underlying sympathetic regeneration to develop therapeutics that can enhance the regeneration of all axon types.
Collapse
|
22
|
Al-Zamil M, Kulikova NG, Shnayder NA, Korchazhkina NB, Petrova MM, Mansur N, Smekalkina LV, Babochkina ZM, Vasilyeva ES, Zhhelambekov IV. Spatial Distribution Dynamics of Sensory Disturbances in the Treatment of Obesity-Related Meralgia Paresthetica Using Transcutaneous Electrical Nerve Stimulation. J Clin Med 2025; 14:390. [PMID: 39860396 PMCID: PMC11765708 DOI: 10.3390/jcm14020390] [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: 11/29/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Background: To date, there have been no studies on the dynamics of areas of pain, paraesthesia and hypoesthesia after the use of various transcutaneous electrical nerve stimulation in the treatment of meralgia paresthetica. Methods: In this pilot study, we observed 68 patients with obesity-related bilateral meralgia paresthetica. Pain syndrome, paraesthesia symptoms, and hypoesthesia were evaluated using 10-point scores. In addition, pain drawing (PD) was used to determine the area of the spatial distribution of pain syndrome and paraesthesia symptoms, and body drawing was used to determine the area of hypoesthesia. Sham TENS was performed in the control group, and effective TENS was performed in the treatment group. The treatment group consisted of two subgroups. One subgroup underwent HF-LA TENS, and the second subgroup underwent LF-HA TENS. Results: Despite the greatest analgesic effect observed from HF-LA TENS, which was assessed using scoring methods, during and after treatment, the reduction in the area of pain and paraesthesia symptoms and the area of hypoesthesia was moderate, short-term, and reversible. In contrast, LF-HA TENS had a pronounced analgesic and sustained anti-paraesthesia effect, manifested by a noticeable decrease in pain and paraesthesia symptoms area in PD, gradually increasing during the first 2 months of follow-up and accompanied by an irreversible prolonged decrease in the area of hypoesthesia. Conclusion: The areas of paraesthesia and hypoesthesia correlate with affective reactions to long-term chronic pain, which noticeably regress under the influence of LF-HA TENS compared to HF-LA TENS.
Collapse
Affiliation(s)
- Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
- Department of Restorative Medicine and Neurorehabilitation, Medical Dental Institute, 127253 Moscow, Russia; (Z.M.B.); (I.V.Z.)
| | - Natalia G. Kulikova
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
- Department of Sports Medicine and Medical Rehabilitation, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Shared Core Facilities “Molecular and Cell Technologies”, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia;
| | - Natalia B. Korchazhkina
- Department of Restorative Medicine and Biomedical Technologies, Federal State Educational Institution of Higher Education, Moscow State University of Medicine and Dentistry Named After A.I. Evdokimov, Ministry of Health of Russia, 127473 Moscow, Russia; (N.B.K.); (E.S.V.)
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, Professor V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia;
| | - Numman Mansur
- City Clinical Hospital Named After V.V. Vinogradov, 117292 Moscow, Russia;
| | - Larisa V. Smekalkina
- Department of Sports Medicine and Medical Rehabilitation, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Zarina M. Babochkina
- Department of Restorative Medicine and Neurorehabilitation, Medical Dental Institute, 127253 Moscow, Russia; (Z.M.B.); (I.V.Z.)
| | - Ekaterina S. Vasilyeva
- Department of Restorative Medicine and Biomedical Technologies, Federal State Educational Institution of Higher Education, Moscow State University of Medicine and Dentistry Named After A.I. Evdokimov, Ministry of Health of Russia, 127473 Moscow, Russia; (N.B.K.); (E.S.V.)
| | - Ivan V. Zhhelambekov
- Department of Restorative Medicine and Neurorehabilitation, Medical Dental Institute, 127253 Moscow, Russia; (Z.M.B.); (I.V.Z.)
| |
Collapse
|
23
|
Redolfi Riva E, Özkan M, Stellacci F, Micera S. Combining external physical stimuli and nanostructured materials for upregulating pro-regenerative cellular pathways in peripheral nerve repair. Front Cell Dev Biol 2024; 12:1491260. [PMID: 39568507 PMCID: PMC11576468 DOI: 10.3389/fcell.2024.1491260] [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: 09/04/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Peripheral nerve repair remains a major clinical challenge, particularly in the pursuit of therapeutic approaches that ensure adequate recovery of patient's activity of daily living. Autografts are the gold standard in clinical practice for restoring lost sensorimotor functions nowadays. However, autografts have notable drawbacks, including dimensional mismatches and the need to sacrifice one function to restore another. Engineered nerve guidance conduits have therefore emerged as promising alternatives. While these conduits show surgical potential, their clinical use is currently limited to the repair of minor injuries, as their ability to reinnervate limiting gap lesions is still unsatisfactory. Therefore, improving patient functional recovery requires a deeper understanding of the cellular mechanisms involved in peripheral nerve regeneration and the development of therapeutic strategies that can precisely modulate these processes. Interest has grown in the use of external energy sources, such as light, ultrasound, electrical, and magnetic fields, to activate cellular pathways related to proliferation, differentiation, and migration. Recent research has explored combining these energy sources with tailored nanostructured materials as nanotransducers to enhance selectivity towards the target cells. This review aims to present the recent findings on this innovative strategy, discussing its potential to support nerve regeneration and its viability as an alternative to autologous transplantation.
Collapse
Affiliation(s)
- Eugenio Redolfi Riva
- Department of Excellence in Robotics and AI, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Melis Özkan
- Bertarelli Foundation Chair in Translational Neural Engineering, Center for Neuroprosthetics and Institute of Bioengineering, École Polytechnique Federale de Lausanne, Lausanne, Switzerland
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Bioengineering and Global Health Institute, Institute of Materials, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Silvestro Micera
- Department of Excellence in Robotics and AI, The BioRobotics Institute, Scuola Superiore Sant'Anna, Pisa, Italy
- Bertarelli Foundation Chair in Translational Neural Engineering, Center for Neuroprosthetics and Institute of Bioengineering, École Polytechnique Federale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
24
|
Vasas NC, Forrest AM, Meyers NA, Christensen MB, Pierce JL, Kaufmann SM, Lanaghen KB, Paniello RC, Barkmeier‐Kraemer JM, Vande Geest JP. A finite element model for biomechanical characterization of ex vivo peripheral nerve dysfunction during stretch. Physiol Rep 2024; 12:e70125. [PMID: 39537361 PMCID: PMC11560341 DOI: 10.14814/phy2.70125] [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: 06/28/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Peripheral nerve damage can cause debilitating symptoms ranging from numbness and pain to sensory loss and atrophy. To uncover the underlying mechanisms of peripheral nerve injury, our research aims to develop a relationship between biomechanical peripheral nerve damage and function through finite element modeling. A noncontact, ex vivo electrophysiology chamber, capable of axially stretching explanted nerves while recording electrical signals, was used to investigate peripheral nerve injury. Successive stretch trials were run on eight sciatic nerves (four females and four males) excised from Sprague-Dawley rats. Nerves were stretched until 50% compound action potential (CAP) amplitude reduction was obtained. A constitutive model developed by Raghavan and Vorp was suitable for rat sciatic nerves, with an average α and β of 0.183 MPa and 1.88 MPa, respectively. We then generated 95% confidence intervals for the stretch at which specific CAP amplitude reductions would occur, which compares well to previous studies. We also developed a finite element model that can predict stretch-induced signaling deficits, applicable for complex nerve geometries and injuries. This relationship between nerve biomechanics and function can be expanded upon to create a clinical model for peripheral nerve dysfunction due to stretch.
Collapse
Affiliation(s)
- Nicholas C. Vasas
- Department of Bioengineering, Swanson School of EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Adam M. Forrest
- Department of Bioengineering, Swanson School of EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Nathaniel A. Meyers
- Department of Bioengineering, Swanson School of EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Michael B. Christensen
- Department of Otolaryngology – Head & Neck SurgeryUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Division of Urology, Department of SurgeryUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Jenny L. Pierce
- Department of Otolaryngology – Head & Neck SurgeryUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Sidney M. Kaufmann
- Department of Otolaryngology – Head & Neck SurgeryUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Kimberly B. Lanaghen
- Department of Otolaryngology – Head & Neck SurgeryUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Randal C. Paniello
- Department of Otolaryngology–Head and Neck SurgeryWashington University School of MedicineSt. LouisMissouriUSA
| | - Julie M. Barkmeier‐Kraemer
- Department of Otolaryngology – Head & Neck SurgeryUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Jonathan P. Vande Geest
- Department of Bioengineering, Swanson School of EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
- McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
25
|
Yu B, Bai J, Guan Y, Huang X, Liang L, Ren Z, Song X, Zhang T, Yang C, Dai F, Wang X, Sheng X, Peng J, Wang L, Wang Y, Yin L. Fully biodegradable and self-powered nerve guidance conduit based on zinc-molybdenum batteries for peripheral nerve repair. Biosens Bioelectron 2024; 263:116578. [PMID: 39038398 DOI: 10.1016/j.bios.2024.116578] [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: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Peripheral nerve injury (PNI) poses a significant public health issue, often leading to muscle atrophy and persistent neuropathic pain, which can drastically impact the quality of life for patients. Electrical stimulation represents an effective and non-pharmacological treatment to promote nerve regeneration. Yet, the postoperative application of electrical stimulation remains a challenge. Here, we propose a fully biodegradable, self-powered nerve guidance conduit (NGC) based on dissolvable zinc-molybdenum batteries. The conduit can offer topographic guidance for nerve regeneration and deliver sustained electrical cues between both ends of a transected nerve stump, extending beyond the surgical window. Schwann cell proliferation and adenosine triphosphate (ATP) production are enhanced by the introduction of the zinc-molybdenum batteries. In rodent models with 10-mm sciatic nerve damage, the device effectively enhances nerve regeneration and motor function recovery. This study offers innovative strategies for creating biodegradable and electroactive devices that hold important promise to optimize therapeutic outcomes for nerve regeneration.
Collapse
Affiliation(s)
- Bingbing Yu
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Jun Bai
- 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 & Injuries PLA, Beijing, 100048, China; Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, 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 & Injuries PLA, Beijing, 100048, China; Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Xueying Huang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Lijing Liang
- 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 & Injuries PLA, Beijing, 100048, China
| | - Zhiqi Ren
- 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 & Injuries PLA, Beijing, 100048, China
| | - Xiangyu Song
- 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 & Injuries PLA, Beijing, 100048, China; Hebei North University, Zhangjiakou, 075051, China
| | - Tieyuan Zhang
- 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 & Injuries PLA, Beijing, 100048, China; Shandong University Center for Orthopedics, Cheeloo College of Medicine, Shandong University, Jinan, 250012, 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, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Fanqi Dai
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Xibo Wang
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Laboratory of Flexible Electronics Technology, 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 & Injuries PLA, Beijing, 100048, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, 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, 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 & Injuries PLA, Beijing, 100048, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, China.
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
26
|
Hashim HT, Varney J, Qais Z, Reda A, Qaderi S, Chowdhury NS, Murry K, Shah J, Alhaideri A, Ahmad S, Hashim AT, Rehman R, Ahmed N, Al-Jorani MS, Skuk M, Abdalhusain M, Khalafalla K. Direct and Gradual Electrical Testicular Shocks Stimulate Spermatogenesis and Activate Sperms in Infertile Men: A Randomized Controlled Trial. Am J Mens Health 2024; 18:15579883241296881. [PMID: 39601214 PMCID: PMC11603473 DOI: 10.1177/15579883241296881] [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: 08/02/2024] [Revised: 08/21/2024] [Accepted: 10/13/2024] [Indexed: 11/29/2024] Open
Abstract
Infertility was reported in approximately 15% of all heterozygous couples, with the male factor accounting for nearly half of the cases. This typically occurs due to low sperm production, sperm dysfunction, and sperm delivery obstruction. In this randomized controlled single-blind clinical trial, 90 infertile male subjects diagnosed with oligospermia, hypospermia, asthenozoospermia, or necrozoospermia were recruited. Semen samples were obtained with the masturbation method and an assessment of semen volume, sperm count, and motility was performed. Five milliamps of electrical shock was delivered to the participants through the fertility improvement device. Semen analysis was collected 4 months post-intervention from all subjects. Data were collected and an analysis of pre- and post-intervention results was performed. There was an improvement in the count, volume, and motility of the patient's sperm after electrical shock treatment compared with the control group. By using the analysis of variance (ANOVA) test, there were statistically significant differences between the first and the second seminal analysis results (<.05). All other results were found to be independently correlated. This study demonstrated that using a painless, convenient at-home device, which is designed to contain all the testis tissue as a cup and then extend to include the scrotal roots reaching the penile root to include the epididymis, could significantly improve sperm motility and count. This device can be utilized to tackle the significant issue of infertility in a cost-effective, safe, and efficacious manner. An ultrasound was done before and after using the device as well as years after with no changes noted.Clinical Trial's Registration Number: NCT04173052.
Collapse
Affiliation(s)
| | - Joseph Varney
- American University of the Caribbean School of Medicine, Cupecoy, Sint Maarten
| | - Zanyar Qais
- Medical University of Lublin, Lublin, Poland
| | - Abdallah Reda
- Faculty of General Medicine, University of Medicine and Pharmacy “Carol Davila” Bucharest, Bucharest, Romania
| | - Shohra Qaderi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | | | | | - Jaffer Shah
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Adil Alhaideri
- College of Medicine, University of Baghdad, Baghdad, Iraq
| | | | | | | | | | | | - Moatamn Skuk
- Al-Kindy Teaching Hospital, Al-Kindy College of Medicine, Baghdad, Iraq
| | | | - Kareim Khalafalla
- Urology Department, McGovern Medical School, University of Texas, Houston, TX, USA
- Urology Department, MD Anderson Cancer Center, Houston, TX, USA
- Urology Department, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
27
|
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
|
28
|
Preetam S, Ghosh A, Mishra R, Pandey A, Roy DS, Rustagi S, Malik S. Electrical stimulation: a novel therapeutic strategy to heal biological wounds. RSC Adv 2024; 14:32142-32173. [PMID: 39399261 PMCID: PMC11467653 DOI: 10.1039/d4ra04258a] [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] [Received: 06/11/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Electrical stimulation (ES) has emerged as a powerful therapeutic modality for enhancing biological wound healing. This non-invasive technique utilizes low-level electrical currents to promote tissue regeneration and expedite the wound healing process. ES has been shown to accelerate wound closure, reduce inflammation, enhance angiogenesis, and modulate cell migration and proliferation through various mechanisms. The principle goal of wound management is the rapid recovery of the anatomical continuity of the skin, to prevent infections from the external environment and maintain homeostasis conditions inside. ES at the wound site is a compelling strategy for skin wound repair. Several ES applications are described in medical literature like AC, DC, and PC to improve cutaneous perfusion and accelerate wound healing. This review aimed to evaluate the primary factors and provides an overview of the potential benefits and mechanisms of ES in wound healing, and its ability to stimulate cellular responses, promote tissue regeneration, and improve overall healing outcomes. We also shed light on the application of ES which holds excellent promise as an adjunct therapy for various types of wounds, including chronic wounds, diabetic ulcers, and surgical incisions.
Collapse
Affiliation(s)
- Subham Preetam
- Department of Robotics and Mechatronics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu 42988 Republic of Korea
| | - Arka Ghosh
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology Bhubaneswar 751003 Odisha India
| | - Richa Mishra
- Department of Computer Engineering, Parul Institute of Engineering and Technology (PIET), Parul University Ta. Waghodia Vadodara Gujarat 391760 India
| | - Arunima Pandey
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology Bhubaneswar 751003 Odisha India
| | - Debanjan Singha Roy
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology Bhubaneswar 751003 Odisha India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University 22 Dehradun Uttarakhand India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand Ranchi Jharkhand 834001 India
- Department of Biotechnology, University Center for Research & Development (UCRD) Chandigarh University Ludhiana Highway Mohali 140413 Punjab India
| |
Collapse
|
29
|
Wu W, Zhang J, Chen Y, Chen Q, Liu Q, Zhang F, Li S, Wang X. Genes in Axonal Regeneration. Mol Neurobiol 2024; 61:7431-7447. [PMID: 38388774 DOI: 10.1007/s12035-024-04049-z] [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: 09/13/2023] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
This review explores the molecular and genetic underpinnings of axonal regeneration and functional recovery post-nerve injury, emphasizing its significance in reversing neurological deficits. It presents a systematic exploration of the roles of various genes in axonal regrowth across peripheral and central nerve injuries. Initially, it highlights genes and gene families critical for axonal growth and guidance, delving into their roles in regeneration. It then examines the regenerative microenvironment, focusing on the role of glial cells in neural repair through dedifferentiation, proliferation, and migration. The concept of "traumatic microenvironments" within the central nervous system (CNS) and peripheral nervous system (PNS) is discussed, noting their impact on regenerative capacities and their importance in therapeutic strategy development. Additionally, the review delves into axonal transport mechanisms essential for accurate growth and reinnervation, integrating insights from proteomics, genome-wide screenings, and gene editing advancements. Conclusively, it synthesizes these insights to offer a comprehensive understanding of axonal regeneration's molecular orchestration, aiming to inform effective nerve injury therapies and contribute to regenerative neuroscience.
Collapse
Affiliation(s)
- Wenshuang Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Jing Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Qianqian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Qianyan Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Fuchao Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Shiying Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
30
|
Brown RI, Barber HM, Kucenas S. Satellite glial cell manipulation prior to axotomy enhances developing dorsal root ganglion central branch regrowth into the spinal cord. Glia 2024; 72:1766-1784. [PMID: 39141572 PMCID: PMC11325082 DOI: 10.1002/glia.24581] [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: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 08/16/2024]
Abstract
The central and peripheral nervous systems (CNS and PNS, respectively) exhibit remarkable diversity in the capacity to regenerate following neuronal injury with PNS injuries being much more likely to regenerate than those that occur in the CNS. Glial responses to damage greatly influence the likelihood of regeneration by either promoting or inhibiting axonal regrowth over time. However, despite our understanding of how some glial lineages participate in nerve degeneration and regeneration, less is known about the contributions of peripheral satellite glial cells (SGC) to regeneration failure following central axon branch injury of dorsal root ganglia (DRG) sensory neurons. Here, using in vivo, time-lapse imaging in larval zebrafish coupled with laser axotomy, we investigate the role of SGCs in axonal regeneration. In our studies we show that SGCs respond to injury by relocating their nuclei to the injury site during the same period that DRG neurons produce new central branch neurites. Laser ablation of SGCs prior to axon injury results in more neurite growth attempts and ultimately a higher rate of successful central axon regrowth, implicating SGCs as inhibitors of regeneration. We also demonstrate that this SGC response is mediated in part by ErbB signaling, as chemical inhibition of this receptor results in reduced SGC motility and enhanced central axon regrowth. These findings provide new insights into SGC-neuron interactions under injury conditions and how these interactions influence nervous system repair.
Collapse
Affiliation(s)
- Robin I Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Heather M Barber
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
- Cell & Developmental Biology Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
31
|
Huang J, Wang P, Wang W, Wei J, Yang L, Liu Z, Li G. Using Electrical Muscle Stimulation to Enhance Electrophysiological Performance of Agonist-Antagonist Myoneural Interface. Bioengineering (Basel) 2024; 11:904. [PMID: 39329646 PMCID: PMC11444137 DOI: 10.3390/bioengineering11090904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The agonist-antagonist myoneural interface (AMI), a surgical method to reinnervate physiologically-relevant proprioceptive feedback for control of limb prostheses, has demonstrated the ability to provide natural afferent sensations for limb amputees when actuating their prostheses. Following AMI surgery, one potential challenge is atrophy of the disused muscles, which would weaken the reinnervation efficacy of AMI. It is well known that electrical muscle stimulus (EMS) can reduce muscle atrophy. In this study, we conducted an animal investigation to explore whether the EMS can significantly improve the electrophysiological performance of AMI. AMI surgery was performed in 14 rats, in which the distal tendons of bilateral solei donors were connected and positioned on the surface of the left biceps femoris. Subsequently, the left tibial nerve and the common peroneus nerve were sutured onto the ends of the connected donor solei. Two stimulation electrodes were affixed onto the ends of the donor solei for EMS delivery. The AMI rats were randomly divided into two groups. One group received the EMS treatment (designated as EMS_on) regularly for eight weeks and another received no EMS (designated as EMS_off). Two physiological parameters, nerve conduction velocity (NCV) and motor unit number, were derived from the electrically evoked compound action potential (CAP) signals to assess the electrophysiological performance of AMI. Our experimental results demonstrated that the reinnervated muscles of the EMS_on group generated higher CAP signals in comparison to the EMS_off group. Both NCV and motor unit number were significantly elevated in the EMS_on group. Moreover, the EMS_on group displayed statistically higher CAP signals on the indirectly activated proprioceptive afferents than the EMS_off group. These findings suggested that EMS treatment would be promising in enhancing the electrophysiological performance and facilitating the reinnervation process of AMI.
Collapse
Affiliation(s)
- Jianping Huang
- Shenzhen Institute of Advanced Technology of the Chinese Academy of Sciences, Shenzhen 518055, China; (J.H.); (W.W.); (J.W.); (L.Y.)
- CAS Key Laboratory of Human-Machine Intelligence-Synergy Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100864, China
| | - Ping Wang
- Biomedical Sensing Engineering and Technology Research Center, Shandong University, Jinan 250000, China;
| | - Wei Wang
- Shenzhen Institute of Advanced Technology of the Chinese Academy of Sciences, Shenzhen 518055, China; (J.H.); (W.W.); (J.W.); (L.Y.)
| | - Jingjing Wei
- Shenzhen Institute of Advanced Technology of the Chinese Academy of Sciences, Shenzhen 518055, China; (J.H.); (W.W.); (J.W.); (L.Y.)
| | - Lin Yang
- Shenzhen Institute of Advanced Technology of the Chinese Academy of Sciences, Shenzhen 518055, China; (J.H.); (W.W.); (J.W.); (L.Y.)
| | - Zhiyuan Liu
- Shenzhen Institute of Advanced Technology of the Chinese Academy of Sciences, Shenzhen 518055, China; (J.H.); (W.W.); (J.W.); (L.Y.)
- Biomedical Sensing Engineering and Technology Research Center, Shandong University, Jinan 250000, China;
| | - Guanglin Li
- Shenzhen Institute of Advanced Technology of the Chinese Academy of Sciences, Shenzhen 518055, China; (J.H.); (W.W.); (J.W.); (L.Y.)
- CAS Key Laboratory of Human-Machine Intelligence-Synergy Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100864, China
- The SIAT Branch, Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen 518055, China
- Shandong Zhongke Advanced Technology Co., Ltd., Jinan 250000, China
| |
Collapse
|
32
|
Bai H, Zhang S, Yang H, Wang J, Chen H, Li J, Li L, Yang Q, Peng B, Zhu Z, Ni S, Liu K, Lei W, Tao TH, Feng Y. Advanced nerve regeneration enabled by neural conformal electronic stimulators enhancing mitochondrial transport. Bioact Mater 2024; 39:287-301. [PMID: 38827170 PMCID: PMC11143791 DOI: 10.1016/j.bioactmat.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/04/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024] Open
Abstract
Addressing peripheral nerve defects remains a significant challenge in regenerative neurobiology. Autografts emerged as the gold-standard management, however, are hindered by limited availability and potential neuroma formation. Numerous recent studies report the potential of wireless electronic system for nerve defects repair. Unfortunately, few has met clinical needs for inadequate electrode precision, poor nerve entrapment and insufficient bioactivity of the matrix material. Herein, we present an advanced wireless electrical nerve stimulator, based on water-responsive self-curling silk membrane with excellent bioabsorbable and biocompatible properties. We constructed a unique bilayer structure with an oriented pre-stretched inner layer and a general silk membrane as outer layer. After wetting, the simultaneous contraction of inner layer and expansion of outer layer achieved controllable super-contraction from 2D flat surface to 3D structural reconfiguration. It enables shape-adaptive wrapping to cover around nerves, overcomes the technical obstacle of preparing electrodes on the inner wall of the conduit, and prevents electrode breakage caused by material expansion in water. The use of fork capacitor-like metal interface increases the contact points between the metal and the regenerating nerve, solving the challenge of inefficient and rough electrical stimulation methods in the past. Newly developed electronic stimulator is effective in restoring 10 mm rat sciatic nerve defects comparable to autologous grafts. The underlying mechanism involves that electric stimulation enhances anterograde mitochondrial transport to match energy demands. This newly introduced device thereby demonstrated the potential as a viable and efficacious alternative to autografts for enhancing peripheral nerve repair and functional recovery.
Collapse
Affiliation(s)
- Hao Bai
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Siqi Zhang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Huiran Yang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hongli Chen
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, Fujian, China
| | - Qian Yang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, PR China
| | - Ziyi Zhu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Siyuan Ni
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Keyin Liu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tiger H. Tao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 200031, China
- Institute of Brain-Intelligence Technology, Zhangjiang Laboratory, Shanghai, 200031, China
| | - Yafei Feng
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
33
|
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
|
34
|
Smadi BM, Shekouhi R, Azizi A, Chim H. Development of Biomaterials for Addressing Upper Extremity Peripheral Nerve Gaps. JOURNAL OF HAND SURGERY GLOBAL ONLINE 2024; 6:711-717. [PMID: 39381386 PMCID: PMC11456663 DOI: 10.1016/j.jhsg.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 10/10/2024] Open
Abstract
Peripheral nerve injuries within the upper extremities can lead to impaired function and reduced quality of life. Although autografts have traditionally served as the primary therapeutic approach to bridge nerve gaps, these present challenges related to donor site morbidity. This review delves into the realm of biomaterials tailored for addressing nerve gaps. Biomaterials, whether natural or synthetically derived, offer the potential not only to act as scaffolds for nerve regeneration but also to be enhanced with growth factors and agents that promote nerve recovery. The historical progression of these biomaterials as well as their current applications, advantages, inherent challenges, and future impact in the arena of regenerative medicine are discussed. By providing a comprehensive overview, we aim to shed light on the transformative potential of biomaterials in peripheral nerve repair and the path toward refining their efficacy in clinical settings.
Collapse
Affiliation(s)
- Bassam M. Smadi
- J Crayton Pruitt Department of Biomedical Engineering, University of Florida, Gainesville, FL
- Nanoscience Institute for Medical and Engineering Technology (NIMET), University of Florida, Gainesville, FL
- College of Medicine, University of Florida, Gainesville, FL
| | - Ramin Shekouhi
- Division of Plastic & Reconstructive Surgery, Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Armina Azizi
- Division of Plastic & Reconstructive Surgery, Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| | - Harvey Chim
- Division of Plastic & Reconstructive Surgery, Department of Surgery, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
35
|
Poitras TM, Komirishetty P, Areti A, Larouche M, Krishnan A, Chandrasekhar A, Munchrath E, Zochodne DW. Manipulation of the Myc Interactome to Enhance Nerve Regeneration in a Murine Model. Ann Neurol 2024; 96:216-230. [PMID: 38818756 DOI: 10.1002/ana.26950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE This study was undertaken to explore manipulation of the Myc protein interactome, members of an oncogene group, in enhancing the intrinsic growth of injured peripheral adult postmitotic neurons and the nerves they supply. New approaches to enhance adult neuron growth properties are a key strategy in improving nerve regeneration. METHODS Expression and impact of Myc interactome members c-Myc, N-Myc, Mad1, and Max were evaluated within naive and "preconditioned" adult sensory neurons and Schwann cells (SCs), using siRNA and transfection of CRISPR/Cas9 or luciferase reporter in vitro. Morphological, behavioral, and electrophysiological indices of nerve regeneration were analyzed in vivo. RESULTS c-Myc, N-Myc, Max, and Mad were expressed in adult sensory neurons and in partnering SCs. In vitro knockdown (KD) of either Mad1 or Max, competitive inhibitors of Myc, unleashed heightened neurite outgrowth in both naive uninjured or preconditioned adult neurons. In contrast, KD or inhibition of both isoforms of Myc was required to suppress growth. In SCs, Mad1 KD not only enhanced migratory behavior but also conditioned increased outgrowth in separately cultured adult sensory neurons. In vivo, local Mad1 KD improved electrophysiological, behavioral, and structural indices of nerve regeneration out to 60 days of follow-up. INTERPRETATION Members of the Myc interactome, specifically Mad1, are novel targets for improving nerve regeneration. Unleashing of Myc growth signaling through Mad1 KD enhances the regrowth of both peripheral neurons and SCs to facilitate better regrowth of nerves. ANN NEUROL 2024;96:216-230.
Collapse
Affiliation(s)
- Trevor M Poitras
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Prashanth Komirishetty
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aparna Areti
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Matt Larouche
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Anand Krishnan
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Ambika Chandrasekhar
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Easton Munchrath
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
36
|
Duarte VN, Lam VT, Rimicci DS, Thompson-Peer KL. Calcium plays an essential role in early-stage dendrite injury detection and regeneration. Prog Neurobiol 2024; 239:102635. [PMID: 38825174 PMCID: PMC11305834 DOI: 10.1016/j.pneurobio.2024.102635] [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: 11/08/2023] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Dendrites are injured in a variety of clinical conditions such as traumatic brain and spinal cord injuries and stroke. How neurons detect injury directly to their dendrites to initiate a pro-regenerative response has not yet been thoroughly investigated. Calcium plays a critical role in the early stages of axonal injury detection and is also indispensable for regeneration of the severed axon. Here, we report cell and neurite type-specific differences in laser injury-induced elevations of intracellular calcium levels. Using a human KCNJ2 transgene, we demonstrate that hyperpolarizing neurons only at the time of injury dampens dendrite regeneration, suggesting that inhibition of injury-induced membrane depolarization (and thus early calcium influx) plays a role in detecting and responding to dendrite injury. In exploring potential downstream calcium-regulated effectors, we identify L-type voltage-gated calcium channels, inositol triphosphate signaling, and protein kinase D activity as drivers of dendrite regeneration. In conclusion, we demonstrate that dendrite injury-induced calcium elevations play a key role in the regenerative response of dendrites and begin to delineate the molecular mechanisms governing dendrite repair.
Collapse
Affiliation(s)
- Vinicius N Duarte
- Dept of Developmental and Cell Biology, University of California, Irvine, United States
| | - Vicky T Lam
- Dept of Developmental and Cell Biology, University of California, Irvine, United States
| | - Dario S Rimicci
- Dept of Developmental and Cell Biology, University of California, Irvine, United States
| | - Katherine L Thompson-Peer
- Dept of Developmental and Cell Biology, University of California, Irvine, United States; Center for the Neurobiology of Learning and Memory, Irvine, CA, United States; Sue and Bill Gross Stem Cell Research Center, Irvine, CA, United States; Reeve-Irvine Research Center, Irvine, CA, United States.
| |
Collapse
|
37
|
Gordon T. Physiology of Nerve Regeneration: Key Factors Affecting Clinical Outcomes. Hand Clin 2024; 40:337-345. [PMID: 38972678 DOI: 10.1016/j.hcl.2024.03.001] [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
Functional recovery after peripheral nerve injuries is disappointing despite surgical advances in nerve repair. This review summarizes the relatively short window of opportunity for successful nerve regeneration due to the decline in the expression of growth-associated genes and in turn, the decline in regenerative capacity of the injured neurons and the support provided by the denervated Schwann cells, and the atrophy of denervated muscles. Brief, low-frequency electrical stimulation and post-injury exercise regimes ameliorate these deficits in animal models and patients, but the misdirection of regenerating nerve fibers compromises functional recovery and remains an important area of future research.
Collapse
Affiliation(s)
- Tessa Gordon
- Department of Surgery, University of Toronto, Toronto, Ontario M5G 1X8, Canada.
| |
Collapse
|
38
|
Fakhraei Khosravieh Z, Nekounam H, Asgari F, Haghighipour N. Electrospun PAN/PANI/CNT scaffolds and electrical pulses: a pathway to stem cell-derived nerve regeneration. Biomed Phys Eng Express 2024; 10:055010. [PMID: 38959871 DOI: 10.1088/2057-1976/ad5e84] [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: 04/24/2024] [Accepted: 07/03/2024] [Indexed: 07/05/2024]
Abstract
Biocompatible polymer-based scaffolds hold great promise for neural repair, especially when they are coupled with electrostimulation to induce neural differentiation. In this study, a combination of polyacrylonitrile/polyaniline (PAN/PANI) and Carbon Nanotubes (CNTs) were used to fabricate three different biomimetic electrospun scaffolds (samples 1, 2 and 3 containing 0.26 wt%, 1 wt% and 2 wt% of CNTs, respectively). These scaffolds underwent thorough characterization for assessing electroconductivity, tensile strength, wettability, degradability, swelling, XRD, and FTIR data. Notably, scanning electron microscopy (SEM) images revealed a three-dimensional scaffold morphology with aligned fibers ranging from 60 nm to 292 nm in diameter. To comprehensively investigate the impact of electrical stimulation on the nervous differentiation of the stem cells seeded on these scaffolds, cell morphology and adhesion were assessed based on SEM images. Additionally, scaffold biocompatibility was studied through MTT assay. Importantly, Real-Time PCR results indicated the expression of neural markers-Nestin,β-tubulin III, and MAP2-by the cells cultured on these samples. In comparison with the control group, samples 1 and 2 exhibited significant increases in Nestin marker expression, indicating early stages of neuronal differentiation, whileβ-tubulin III expression was significantly reduced and MAP2 expression remained statistically unchanged. In contrast, sample 3 did not display a statistically significant upturn in Nestin maker expression, while showcasing remarkable increases in the expression of both MAP2 andβ-tubulin III, as markers of the end stages of differentiation, leading to postmitotic neurons. These results could be attributed to the higher electroconductivity of S3 compared to other samples. Our findings highlight the biomimetic potential of the prepared scaffolds for neural repair, illustrating their effectiveness in guiding stem cell differentiation toward a neural lineage.
Collapse
Affiliation(s)
| | - Houra Nekounam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Asgari
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
39
|
Choi JH, Moon J, Park YH, Eom K. Computational analysis of electrode structure and configuration for efficient and localized neural stimulation. Biomed Eng Lett 2024; 14:717-726. [PMID: 38946826 PMCID: PMC11208352 DOI: 10.1007/s13534-024-00364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/19/2024] [Accepted: 02/18/2024] [Indexed: 07/02/2024] Open
Abstract
Neuromodulation technique using electric stimulation is widely applied in neural prosthesis, therapy, and neuroscience research. Various stimulation techniques have been developed to enhance stimulation efficiency and to precisely target the specific area of the brain which involves optimizing the geometry and the configuration of the electrode, stimulation pulse type and shapes, and electrode materials. Although the effects of electrode shape, size, and configuration on the performance of neural stimulation have individually been characterized, to date, there is no integrative investigation of how this factor affects neural stimulation. In this study, we computationally modeled the various types of electrodes with varying shapes, sizes, and configurations and simulated the electric field to calculate the activation function. The electrode geometry is then integratively assessed in terms of stimulation efficiency and stimulation focality. We found that stimulation efficiency is enhanced by making the electrode sharper and smaller. A center-to-vertex distance exceeding 100 µm shows enhanced stimulation efficiency in the bipolar configuration. Additionally, the separation distance of less than 1 mm between the reference and stimulation electrodes exhibits higher stimulation efficiency compared to the monopolar configuration. The region of neurons to be stimulated can also be modified. We found that sharper electrodes can locally activate the neuron. In most cases, except for the rectangular electrode shape with a center-to-vertex distance smaller than 100 µm, the bipolar electrode configuration can locally stimulate neurons as opposed to the monopolar configuration. These findings shed light on the optimal selection of neural electrodes depending on the target applications.
Collapse
Affiliation(s)
- Ji Hoon Choi
- Department of Electronics Engineering, College of Engineering, Pusan National University, Busan, 46241 Republic of Korea
| | - Jeongju Moon
- Department of Electronics Engineering, College of Engineering, Pusan National University, Busan, 46241 Republic of Korea
| | - Young Hoon Park
- Department of Electronics Engineering, College of Engineering, Pusan National University, Busan, 46241 Republic of Korea
| | - Kyungsik Eom
- Department of Electronics Engineering, College of Engineering, Pusan National University, Busan, 46241 Republic of Korea
| |
Collapse
|
40
|
Shi S, Ou X, Du X. Enhanced nerve function recovery in radial nerve palsy patients with humerus shaft fracture: a randomized study of low-frequency pulse electrical stimulation combined with exercise therapy. Front Neurol 2024; 15:1370316. [PMID: 39011357 PMCID: PMC11246844 DOI: 10.3389/fneur.2024.1370316] [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: 01/14/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024] Open
Abstract
Objective To evaluate the effect of low-frequency pulse electrical stimulation plus exercise therapy on nerve function recovery in patients with radial nerve palsy after humerus shaft fracture. Methods A total of 110 patients with humerus shaft fracture and radial nerve injury admitted to our hospital from January 2017 to December 2021 were recruited. They were randomized to receive either conventional exercise therapy (control group) or conventional exercise therapy plus low-frequency pulse electrical stimulation (study group) according to the random number table method, with 55 cases in each. Clinical efficacy, muscle strength recovery, nerve conduction velocity (MCV), amplitude, wrist joint, and elbow joint activities of patients were analyzed and compared. Results Patients with low frequency stimulation (LFS) showed significantly higher treatment effectiveness (89.09%) than those with exercise therapy only (69.09%). The incorporation of LFS with exercise therapy provided more enhancement in the muscle strength of wrist extensor and total finger extensor in patients when compared with a mere exercise intervention, suggesting better muscle function recovery of patients produced by LFS. Moreover, a significant increase in MCV and its amplitude was observed in all included patients, among which those receiving LFS showed a greater escalation of MCV and its amplitude. Following a treatment duration of 6 months, more patients in the LFS cohort were reported to achieve a wrist extension and elbow extension with an angle over 45° than the controls. There was no notable variance in adverse responses noted between the two patient groups. Conclusion In patients afflicted with humerus shaft fracture and radial nerve injury, the amalgamation of exercise therapy with low-frequency pulse electrical stimulation can significantly improve clinical efficacy, promote nerve function, and muscle strength recovery, and features a high safety profile. Relevance to clinical practice The combination of exercise therapy and low-frequency pulsed electrical stimulation can notably improve the promotion of neurologic function and muscle strength recovery in patients with humerus shaft fractures and radial nerve injuries with a high degree of safety.Clinical trial registration:https://www.researchregistry.com, identifier researchregistry9461.
Collapse
Affiliation(s)
- Shaoyan Shi
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xuehai Ou
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaolong Du
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
41
|
Li J, Zhang F, Lyu H, Yin P, Shi L, Li Z, Zhang L, Di CA, Tang P. Evolution of Musculoskeletal Electronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303311. [PMID: 38561020 DOI: 10.1002/adma.202303311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 02/10/2024] [Indexed: 04/04/2024]
Abstract
The musculoskeletal system, constituting the largest human physiological system, plays a critical role in providing structural support to the body, facilitating intricate movements, and safeguarding internal organs. By virtue of advancements in revolutionized materials and devices, particularly in the realms of motion capture, health monitoring, and postoperative rehabilitation, "musculoskeletal electronics" has actually emerged as an infancy area, but has not yet been explicitly proposed. In this review, the concept of musculoskeletal electronics is elucidated, and the evolution history, representative progress, and key strategies of the involved materials and state-of-the-art devices are summarized. Therefore, the fundamentals of musculoskeletal electronics and key functionality categories are introduced. Subsequently, recent advances in musculoskeletal electronics are presented from the perspectives of "in vitro" to "in vivo" signal detection, interactive modulation, and therapeutic interventions for healing and recovery. Additionally, nine strategy avenues for the development of advanced musculoskeletal electronic materials and devices are proposed. Finally, concise summaries and perspectives are proposed to highlight the directions that deserve focused attention in this booming field.
Collapse
Affiliation(s)
- Jia Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853, China
| | - Fengjiao Zhang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Houchen Lyu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853, China
| | - Lei Shi
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853, China
| | - Zhiyi Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853, China
| | - Chong-An Di
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853, China
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853, China
| |
Collapse
|
42
|
Alahmadi A, Abdelsamad Y, Yousef M, Almuhawas F, Hafez A, Alzhrani F, Hagr A. Cochlear Implantation: Long-Term Effect of Early Activation on Electrode Impedance. J Clin Med 2024; 13:3299. [PMID: 38893010 PMCID: PMC11172931 DOI: 10.3390/jcm13113299] [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: 04/13/2024] [Revised: 05/17/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Objectives: The growing adoption of cochlear implants (CIs) necessitates understanding the factors influencing long-term performance and improved outcomes. This work investigated the long-term effect of early activation of CIs on electrode impedance in a large sample of CI users at different time points. Methods: A retrospective study on 915 ears from CI patients who were implanted between 2015 and 2020. According to their CI audio processor activation time, the patients were categorized into early activation (activated 1 day after surgery, n = 481) and classical activation (activated 4 weeks after surgery, n = 434) groups. Then, the impact of the activation times on the electrode impedance values, along the electrode array contacts, at different time points up to two years was studied and analyzed. Results: The early activation group demonstrated lower impedance values across all the electrode array sections compared to the classical activation at 1 month, 1 year, and 2 years post-implantation. At 1 month, early activation was associated with a reduction of 0.34 kΩ, 0.46 kΩ, and 0.37 kΩ in the apical, middle, and basal sections, respectively. These differences persisted at subsequent intervals. Conclusions: Early activation leads to sustained reductions in the electrode impedance compared to classical activation (CA), suggesting that earlier activation might positively affect long-term CI outcomes.
Collapse
Affiliation(s)
- Asma Alahmadi
- King Abdullah Ear Specialist Center (KAESC), King Saud Medical City, King Saud University, Riyadh 11411, Saudi Arabia; (M.Y.); (F.A.); (F.A.); (A.H.)
| | - Yassin Abdelsamad
- Research Department, MED-EL GmbH, Riyadh 11563, Saudi Arabia; (Y.A.); (A.H.)
| | - Medhat Yousef
- King Abdullah Ear Specialist Center (KAESC), King Saud Medical City, King Saud University, Riyadh 11411, Saudi Arabia; (M.Y.); (F.A.); (F.A.); (A.H.)
- Audio Vestibular Unit, ENT Department, Menoufia University, Menoufia 32928, Egypt
| | - Fida Almuhawas
- King Abdullah Ear Specialist Center (KAESC), King Saud Medical City, King Saud University, Riyadh 11411, Saudi Arabia; (M.Y.); (F.A.); (F.A.); (A.H.)
| | - Ahmed Hafez
- Research Department, MED-EL GmbH, Riyadh 11563, Saudi Arabia; (Y.A.); (A.H.)
| | - Farid Alzhrani
- King Abdullah Ear Specialist Center (KAESC), King Saud Medical City, King Saud University, Riyadh 11411, Saudi Arabia; (M.Y.); (F.A.); (F.A.); (A.H.)
| | - Abdulrahman Hagr
- King Abdullah Ear Specialist Center (KAESC), King Saud Medical City, King Saud University, Riyadh 11411, Saudi Arabia; (M.Y.); (F.A.); (F.A.); (A.H.)
| |
Collapse
|
43
|
Das JM, Upadhyay J, Monaghan MG, Borah R. Impact of the Reduction Time-Dependent Electrical Conductivity of Graphene Nanoplatelet-Coated Aligned Bombyx mori Silk Scaffolds on Electrically Stimulated Axonal Growth. ACS APPLIED BIO MATERIALS 2024; 7:2389-2401. [PMID: 38502100 PMCID: PMC11022174 DOI: 10.1021/acsabm.4c00052] [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: 01/12/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024]
Abstract
Graphene-based nanomaterials, renowned for their outstanding electrical conductivity, have been extensively studied as electroconductive biomaterials (ECBs) for electrically stimulated tissue regeneration. However, using eco-friendly reducing agents like l-ascorbic acid (l-Aa) can result in lower conductive properties in these ECBs, limiting their full potential for smooth charge transfer in living tissues. Moreover, creating a flexible biomaterial scaffold using these materials that accurately mimics a specific tissue microarchitecture, such as nerves, poses additional challenges. To address these issues, this study developed a microfibrous scaffold of Bombyx mori (Bm) silk fibroin uniformly coated with graphene nanoplatelets (GNPs) through a vacuum coating method. The scaffold's electrical conductivity was optimized by varying the reduction period using l-Aa. The research systematically investigated how different reduction periods impact scaffold properties, focusing on electrical conductivity and its significance on electrically stimulated axonal growth in PC12 cells. Results showed that a 48 h reduction significantly increased surface electrical conductivity by 100-1000 times compared to a shorter or no reduction process. l-Aa contributed to stabilizing the reduced GNPs, demonstrated by a slow degradation profile and sustained conductivity even after 60 days in a proteolytic environment. β (III) tubulin immunostaining of PC12 cells on varied silk:GNP scaffolds under pulsed electrical stimulation (ES, 50 Hz frequency, 1 ms pulse width, and amplitudes of 100 and 300 mV/cm) demonstrates accelerated axonal growth on scaffolds exhibiting higher conductivity. This is supported by upregulated intracellular Ca2+ dynamics immediately after ES on the scaffolds with higher conductivity, subjected to a prolonged reduction period. The study showcases a sustainable reduction approach using l-Aa in combination with natural Bm silk fibroin to create a highly conductive, mechanically robust, and stable silk:GNP-based aligned fibrous scaffold. These scaffolds hold promise for functional regeneration in electrically excitable tissues such as nerves, cardiac tissue, and muscles.
Collapse
Affiliation(s)
- Jitu Mani Das
- Life
Sciences Division, Institute of Advanced
Study in Science & Technology, Guwahati 781035, India
| | - Jnanendra Upadhyay
- Department
of Physics, Dakshin Kamrup College, Kamrup, Mirza, Assam 781125, India
| | - Michael G. Monaghan
- Department
of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin D2, Ireland
- Advanced
Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons
in Ireland, Dublin D2, Ireland
- Trinity
Centre for Biomedical Engineering, Trinity
College Dublin, Dublin D2, Ireland
- CÚRAM,
Centre for Research in Medical Devices, National University of Ireland, Galway H91 W2TY, Ireland
| | - Rajiv Borah
- Life
Sciences Division, Institute of Advanced
Study in Science & Technology, Guwahati 781035, India
- Department
of Mechanical, Manufacturing and Biomedical Engineering, Trinity College Dublin, Dublin D2, Ireland
- Advanced
Materials and BioEngineering Research (AMBER), Centre at Trinity College Dublin and the Royal College of Surgeons
in Ireland, Dublin D2, Ireland
- Trinity
Centre for Biomedical Engineering, Trinity
College Dublin, Dublin D2, Ireland
| |
Collapse
|
44
|
Wu J, Xue W, Yun Z, Liu Q, Sun X. Biomedical applications of stimuli-responsive "smart" interpenetrating polymer network hydrogels. Mater Today Bio 2024; 25:100998. [PMID: 38390342 PMCID: PMC10882133 DOI: 10.1016/j.mtbio.2024.100998] [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: 11/17/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
In recent years, owing to the ongoing advancements in polymer materials, hydrogels have found increasing applications in the biomedical domain, notably in the realm of stimuli-responsive "smart" hydrogels. Nonetheless, conventional single-network stimuli-responsive "smart" hydrogels frequently exhibit deficiencies, including low mechanical strength, limited biocompatibility, and extended response times. In response, researchers have addressed these challenges by introducing a second network to create stimuli-responsive "smart" Interpenetrating Polymer Network (IPN) hydrogels. The mechanical strength of the material can be significantly improved due to the topological entanglement and physical interactions within the interpenetrating structure. Simultaneously, combining different network structures enhances the biocompatibility and stimulus responsiveness of the gel, endowing it with unique properties such as cell adhesion, conductivity, hemostasis/antioxidation, and color-changing capabilities. This article primarily aims to elucidate the stimulus-inducing factors in stimuli-responsive "smart" IPN hydrogels, the impact of the gels on cell behaviors and their biomedical application range. Additionally, we also offer an in-depth exposition of their categorization, mechanisms, performance characteristics, and related aspects. This review furnishes a comprehensive assessment and outlook for the advancement of stimuli-responsive "smart" IPN hydrogels within the biomedical arena. We believe that, as the biomedical field increasingly demands novel materials featuring improved mechanical properties, robust biocompatibility, and heightened stimulus responsiveness, stimuli-responsive "smart" IPN hydrogels will hold substantial promise for wide-ranging applications in this domain.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wu Xue
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhihe Yun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Qinyi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Xinzhi Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
45
|
Izhiman Y, Esfandiari L. Emerging role of extracellular vesicles and exogenous stimuli in molecular mechanisms of peripheral nerve regeneration. Front Cell Neurosci 2024; 18:1368630. [PMID: 38572074 PMCID: PMC10989355 DOI: 10.3389/fncel.2024.1368630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Peripheral nerve injuries lead to significant morbidity and adversely affect quality of life. The peripheral nervous system harbors the unique trait of autonomous regeneration; however, achieving successful regeneration remains uncertain. Research continues to augment and expedite successful peripheral nerve recovery, offering promising strategies for promoting peripheral nerve regeneration (PNR). These include leveraging extracellular vesicle (EV) communication and harnessing cellular activation through electrical and mechanical stimulation. Small extracellular vesicles (sEVs), 30-150 nm in diameter, play a pivotal role in regulating intercellular communication within the regenerative cascade, specifically among nerve cells, Schwann cells, macrophages, and fibroblasts. Furthermore, the utilization of exogenous stimuli, including electrical stimulation (ES), ultrasound stimulation (US), and extracorporeal shock wave therapy (ESWT), offers remarkable advantages in accelerating and augmenting PNR. Moreover, the application of mechanical and electrical stimuli can potentially affect the biogenesis and secretion of sEVs, consequently leading to potential improvements in PNR. In this review article, we comprehensively delve into the intricacies of cell-to-cell communication facilitated by sEVs and the key regulatory signaling pathways governing PNR. Additionally, we investigated the broad-ranging impacts of ES, US, and ESWT on PNR.
Collapse
Affiliation(s)
- Yara Izhiman
- Esfandiari Laboratory, Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Leyla Esfandiari
- Esfandiari Laboratory, Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Department of Electrical and Computer Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
46
|
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
|
47
|
Gouveia D, Cardoso A, Carvalho C, Oliveira AC, Almeida A, Gamboa Ó, Lopes B, Coelho A, Alvites R, Varejão AS, Maurício AC, Ferreira A, Martins Â. Early Intensive Neurorehabilitation in Traumatic Peripheral Nerve Injury-State of the Art. Animals (Basel) 2024; 14:884. [PMID: 38539981 PMCID: PMC10967370 DOI: 10.3390/ani14060884] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/09/2024] [Accepted: 03/11/2024] [Indexed: 02/24/2025] Open
Abstract
Traumatic nerve injuries are common lesions that affect several hundred thousand humans, as well as dogs and cats. The assessment of nerve regeneration through animal models may provide information for translational research and future therapeutic options that can be applied mutually in veterinary and human medicine, from a One Health perspective. This review offers a hands-on vision of the non-invasive and conservative approaches to peripheral nerve injury, focusing on the role of neurorehabilitation in nerve repair and regeneration. The peripheral nerve injury may lead to hypersensitivity, allodynia and hyperalgesia, with the possibility of joint contractures, decreasing functionality and impairing the quality of life. The question remains regarding how to improve nerve repair with surgical possibilities, but also considering electrical stimulation modalities by modulating sensory feedback, upregulation of BDNF, GFNF, TrKB and adenosine monophosphate, maintaining muscle mass and modulating fatigue. This could be improved by the positive synergetic effect of exercises and physical activity with locomotor training, and other physical modalities (low-level laser therapy, ultrasounds, pulsed electromagnetic fields, electroacupuncture and others). In addition, the use of cell-based therapies is an innovative treatment tool in this field. These strategies may help avoid situations of permanent monoplegic limbs that could lead to amputation.
Collapse
Affiliation(s)
- Débora Gouveia
- Arrábida Veterinary Hospital—Arrábida Animal Rehabilitation Center, 2925-538 Setúbal, Portugal; (D.G.); (A.C.); (C.C.); (A.C.O.); (Â.M.)
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Campo Grande, 1950-396 Lisboa, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Campo Grande, 1749-024 Lisboa, Portugal
| | - Ana Cardoso
- Arrábida Veterinary Hospital—Arrábida Animal Rehabilitation Center, 2925-538 Setúbal, Portugal; (D.G.); (A.C.); (C.C.); (A.C.O.); (Â.M.)
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Campo Grande, 1950-396 Lisboa, Portugal
| | - Carla Carvalho
- Arrábida Veterinary Hospital—Arrábida Animal Rehabilitation Center, 2925-538 Setúbal, Portugal; (D.G.); (A.C.); (C.C.); (A.C.O.); (Â.M.)
| | - Ana Catarina Oliveira
- Arrábida Veterinary Hospital—Arrábida Animal Rehabilitation Center, 2925-538 Setúbal, Portugal; (D.G.); (A.C.); (C.C.); (A.C.O.); (Â.M.)
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Campo Grande, 1950-396 Lisboa, Portugal
| | - António Almeida
- Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisboa, Portugal; (A.A.); (Ó.G.); (A.F.)
- Centro Interdisciplinar—Investigação em Saúde Animal (CIISA), Faculdade de Medicina Veterinária, Av. Universidade Técnica de Lisboa, 1300-477 Lisboa, Portugal
| | - Óscar Gamboa
- Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisboa, Portugal; (A.A.); (Ó.G.); (A.F.)
- Centro Interdisciplinar—Investigação em Saúde Animal (CIISA), Faculdade de Medicina Veterinária, Av. Universidade Técnica de Lisboa, 1300-477 Lisboa, Portugal
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA), Universidade do Porto (UP), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (A.C.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal;
| | - André Coelho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA), Universidade do Porto (UP), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (A.C.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal;
| | - Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA), Universidade do Porto (UP), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (A.C.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal;
- Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Artur Severo Varejão
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal;
- CECAV, Centre for Animal Sciences and Veterinary Studies, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA), Universidade do Porto (UP), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal; (B.L.); (A.C.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no. 228, 4050-313 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal;
| | - António Ferreira
- Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisboa, Portugal; (A.A.); (Ó.G.); (A.F.)
- Centro Interdisciplinar—Investigação em Saúde Animal (CIISA), Faculdade de Medicina Veterinária, Av. Universidade Técnica de Lisboa, 1300-477 Lisboa, Portugal
| | - Ângela Martins
- Arrábida Veterinary Hospital—Arrábida Animal Rehabilitation Center, 2925-538 Setúbal, Portugal; (D.G.); (A.C.); (C.C.); (A.C.O.); (Â.M.)
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Campo Grande, 1950-396 Lisboa, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Campo Grande, 1749-024 Lisboa, Portugal
| |
Collapse
|
48
|
Morrell NT, Dahlberg RK, Scott KL. Electrical Stimulation Use in Upper Extremity Peripheral Nerve Injuries. J Am Acad Orthop Surg 2024; 32:156-161. [PMID: 38109725 DOI: 10.5435/jaaos-d-23-00437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/08/2023] [Indexed: 12/20/2023] Open
Abstract
Peripheral nerve injuries can be debilitating and often have a variable course of recovery. Electrical stimulation (ES) has been used as an intervention to attempt to overcome the limits of peripheral nerve surgery and improve patient outcomes after peripheral nerve injury. Little has been written in the orthopaedic literature regarding the use of this technology. The purpose of this review was to provide a focused analysis of past and current literature surrounding the utilization of ES in the treatment of various upper extremity peripheral nerve pathologies including compression neuropathies and nerve transection. We aimed to provide clarity on the clinical benefits, appropriate timing for its employment, risks and limitations, and the need for future studies of ES.
Collapse
Affiliation(s)
- Nathan T Morrell
- Department of Orthopedics and Rehabilitation, University of New Mexico, Albuquerque, NM (Morrell and Dahlberg), Banner University Medical Center, Glendale, AZ (Scott)
| | | | | |
Collapse
|
49
|
Kong J, Teng C, Liu F, Wang X, Zhou Y, Zong Y, Wan Z, Qin J, Yu B, Mi D, Wang Y. Enhancing regeneration and repair of long-distance peripheral nerve defect injuries with continuous microcurrent electrical nerve stimulation. Front Neurosci 2024; 18:1361590. [PMID: 38406586 PMCID: PMC10885699 DOI: 10.3389/fnins.2024.1361590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Peripheral nerve injuries, especially those involving long-distance deficits, pose significant challenges in clinical repair. This study explores the potential of continuous microcurrent electrical nerve stimulation (cMENS) as an adjunctive strategy to promote regeneration and repair in such cases. Methods The study initially optimized cMENS parameters and assessed its impact on Schwann cell activity, neurotrophic factor secretion, and the nerve regeneration microenvironment. Subsequently, a rat sciatic nerve defect-bridge repair model was employed to evaluate the reparative effects of cMENS as an adjuvant treatment. Functional recovery was assessed through gait analysis, motor function tests, and nerve conduction assessments. Additionally, nerve regeneration and denervated muscle atrophy were observed through histological examination. Results The study identified a 10-day regimen of 100uA microcurrent stimulation as optimal. Evaluation focused on Schwann cell activity and the microenvironment, revealing the positive impact of cMENS on maintaining denervated Schwann cell proliferation and enhancing neurotrophic factor secretion. In the rat model of sciatic nerve defect-bridge repair, cMENS demonstrated superior effects compared to control groups, promoting motor function recovery, nerve conduction, and sensory and motor neuron regeneration. Histological examinations revealed enhanced maturation of regenerated nerve fibers and reduced denervated muscle atrophy. Discussion While cMENS shows promise as an adjuvant treatment for long-distance nerve defects, future research should explore extended stimulation durations and potential synergies with tissue engineering grafts to improve outcomes. This study contributes comprehensive evidence supporting the efficacy of cMENS in enhancing peripheral nerve regeneration.
Collapse
Affiliation(s)
- Junjie Kong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Cheng Teng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fenglan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xuzhaoyu Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yi Zhou
- Department of Orthopedics, Nantong City Hospital of Traditional Chinese Medicine, Nantong, China
| | - Ying Zong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zixin Wan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jun Qin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Daguo Mi
- Department of Orthopedics, Nantong City Hospital of Traditional Chinese Medicine, Nantong, China
| | - Yaxian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Hospital and Medical School, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
50
|
Hu M, Li H, Zhu K, Guo L, Zhao M, Zhan H, Devreotes PN, Qing Q. Electric field modulation of ERK dynamics shows dependency on waveform and timing. Sci Rep 2024; 14:3167. [PMID: 38326365 PMCID: PMC10850077 DOI: 10.1038/s41598-024-53018-y] [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: 09/24/2023] [Accepted: 01/25/2024] [Indexed: 02/09/2024] Open
Abstract
Different exogenous electric fields (EF) can guide cell migration, disrupt proliferation, and program cell development. Studies have shown that many of these processes were initiated at the cell membrane, but the mechanism has been unclear, especially for conventionally non-excitable cells. In this study, we focus on the electrostatic aspects of EF coupling with the cell membrane by eliminating Faradaic processes using dielectric-coated microelectrodes. Our data unveil a distinctive biphasic response of the ERK signaling pathway of epithelial cells (MCF10A) to alternate current (AC) EF. The ERK signal exhibits both inhibition and activation phases, with the former triggered by a lower threshold of AC EF, featuring a swifter peaking time and briefer refractory periods than the later-occurring activation phase, induced at a higher threshold. Interestingly, the biphasic ERK responses are sensitive to the waveform and timing of EF stimulation pulses, depicting the characteristics of electrostatic and dissipative interactions. Blocker tests and correlated changes of active Ras on the cell membrane with ERK signals indicated that both EGFR and Ras were involved in the rich ERK dynamics induced by EF. We propose that the frequency-dependent dielectric relaxation process could be an important mechanism to couple EF energy to the cell membrane region and modulate membrane protein-initiated signaling pathways, which can be further explored to precisely control cell behavior and fate with high temporal and spatial resolution.
Collapse
Affiliation(s)
- Minxi Hu
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Houpu Li
- Department of Physics, Arizona State University, Tempe, AZ, 85287, USA
| | - Kan Zhu
- Department of Dermatology, University of California, Davis, CA, 95616, USA
| | - Liang Guo
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Min Zhao
- Department of Dermatology, University of California, Davis, CA, 95616, USA
- Department of Ophthalmology and Vision Science, University of California, Davis, CA, 95616, USA
| | - Huiwang Zhan
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Peter N Devreotes
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Quan Qing
- Department of Physics, Arizona State University, Tempe, AZ, 85287, USA.
- Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|