1
|
Hao M, Chu J, Zhang T, Yin T, Gu Y, Liang W, Ji W, Zhuang J, Liu Y, Gao J, Yin Y. Nanomaterials-mediated lysosomal regulation: a robust protein-clearance approach for the treatment of Alzheimer's disease. Neural Regen Res 2025; 20:424-439. [PMID: 38819046 PMCID: PMC11317947 DOI: 10.4103/nrr.nrr-d-23-01736] [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: 10/22/2023] [Revised: 01/04/2024] [Accepted: 02/20/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a debilitating, progressive neurodegenerative disorder characterized by the progressive accumulation of abnormal proteins, including amyloid plaques and intracellular tau tangles, primarily within the brain. Lysosomes, crucial intracellular organelles responsible for protein degradation, play a key role in maintaining cellular homeostasis. Some studies have suggested a link between the dysregulation of the lysosomal system and pathogenesis of neurodegenerative diseases, including Alzheimer's disease. Restoring the normal physiological function of lysosomes hold the potential to reduce the pathological burden and improve the symptoms of Alzheimer's disease. Currently, the efficacy of drugs in treating Alzheimer's disease is limited, with major challenges in drug delivery efficiency and targeting. Recently, nanomaterials have gained widespread use in Alzheimer's disease drug research owing to their favorable physical and chemical properties. This review aims to provide a comprehensive overview of recent advances in using nanomaterials (polymeric nanomaterials, nanoemulsions, and carbon-based nanomaterials) to enhance lysosomal function in treating Alzheimer's disease. This review also explores new concepts and potential therapeutic strategies for Alzheimer's disease through the integration of nanomaterials and modulation of lysosomal function. In conclusion, this review emphasizes the potential of nanomaterials in modulating lysosomal function to improve the pathological features of Alzheimer's disease. The application of nanotechnology to the development of Alzheimer's disease drugs brings new ideas and approaches for future treatment of this disease.
Collapse
Affiliation(s)
- Mengqi Hao
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jianjian Chu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tong Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yuankai Gu
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Wendanqi Liang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wenbo Ji
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University (Shanghai Changzheng Hospital), Shanghai, China
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
2
|
Rizoli C, Dos Santos NM, Maróstica Júnior MR, da Cruz-Höfling MA, Mendonça MCP, de Jesus MB. The therapeutic potential of reduced graphene oxide in attenuating cuprizone-induced demyelination in mice. NANOTECHNOLOGY 2024; 36:025102. [PMID: 39389086 DOI: 10.1088/1361-6528/ad857e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024]
Abstract
Reduced graphene oxide (rGO) has unique physicochemical properties that make it suitable for therapeutic applications in neurodegenerative scenarios. This study investigates the therapeutic potential of rGO in a cuprizone-induced demyelination model in mice through histomorphological techniques and analysis of biochemical parameters. We demonstrate that daily intraperitoneal administration of rGO (1 mg ml-1) for 21 days tends to reduce demyelination in theCorpus callosumby decreasing glial cell recruitment during the repair mechanism. Additionally, rGO interferes with oxidative stress markers in the brain and liver indicating potential neuroprotective effects in the central nervous system. No significant damage to vital organs was observed, suggesting that multiple doses could be used safely. However, further long-term investigations are needed to understand rGO distribution, metabolism, routes of action and associated challenges in central neurodegenerative therapies. Overall, these findings contribute to the comprehension of rGO effectsin vivo, paving the way for possible future clinical research.
Collapse
Affiliation(s)
- Cintia Rizoli
- Departmento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | - Maria Alice da Cruz-Höfling
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Marcelo Bispo de Jesus
- Departmento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
3
|
Song J, Dong J, Yuan Z, Huang M, Yu X, Zhao Y, Shen Y, Wu J, El-Newehy M, Abdulhameed MM, Sun B, Chen J, Mo X. Shape-Persistent Conductive Nerve Guidance Conduits for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2401160. [PMID: 38757919 DOI: 10.1002/adhm.202401160] [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: 03/27/2024] [Revised: 05/09/2024] [Indexed: 05/18/2024]
Abstract
To solve the problems of slow regeneration and mismatch of axon regeneration after peripheral nerve injury, nerve guidance conduits (NGCs) have been widely used to promote nerve regeneration. Multichannel NGCs have been widely studied to mimic the structure of natural nerve bundles. However, multichannel conduits are prone to structural instability. Thermo-responsive shape memory polymers (SMPs) can maintain a persistent initial structure over the body temperature range. Electrical stimulation (ES), utilized within nerve NGCs, serves as a biological signal to expedite damaged nerve regeneration. Here, an electrospun shape-persistent conductive NGC is designed to maintain the persistent tubular structure in the physiological temperature range and improve the conductivity. The physicochemical and biocompatibility of these P, P/G, P/G-GO, and P/G-RGO NGCs are conducted in vitro. Meanwhile, to evaluate biocompatibility and peripheral nerve regeneration, NGCs are implanted in subcutaneous parts of the back of rats and sciatic nerves assessed by histology and immunofluorescence analyses. The conductive NGC displays a stable structure, good biocompatibility, and promoted nerve regeneration. Collectively, the shape-persistent conductive NGC (P/G-RGO) is expected to promote peripheral nerve recovery, especially for long-gap and large-diameter nerves.
Collapse
Affiliation(s)
- Jiahui Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Jize Dong
- Department of Sports Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200080, P. R. China
| | - Zhengchao Yuan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Moran Huang
- Department of Sports Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200080, P. R. China
| | - Xiao Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Yue Zhao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Yihong Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Jinglei Wu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Meera Moydeen Abdulhameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Binbin Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Jiwu Chen
- Department of Sports Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, 200080, P. R. China
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| |
Collapse
|
4
|
Kohestani AA, Xu Z, Baştan FE, Boccaccini AR, Pishbin F. Electrically conductive coatings in tissue engineering. Acta Biomater 2024; 186:30-62. [PMID: 39128796 DOI: 10.1016/j.actbio.2024.08.007] [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: 04/14/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Recent interest in tissue engineering (TE) has focused on electrically conductive biomaterials. This has been inspired by the characteristics of the cells' microenvironment where signalling is supported by electrical stimulation. Numerous studies have demonstrated the positive influence of electrical stimulation on cell excitation to proliferate, differentiate, and deposit extracellular matrix. Even without external electrical stimulation, research shows that electrically active scaffolds can improve tissue regeneration capacity. Tissues like bone, muscle, and neural contain electrically excitable cells that respond to electrical cues provided by implanted biomaterials. To introduce an electrical pathway, TE scaffolds can incorporate conductive polymers, metallic nanoparticles, and ceramic nanostructures. However, these materials often do not meet implantation criteria, such as maintaining mechanical durability and degradation characteristics, making them unsuitable as scaffold matrices. Instead, depositing conductive layers on TE scaffolds has shown promise as an efficient alternative to creating electrically conductive structures. A stratified scaffold with an electroactive surface synergistically excites the cells through active top-pathway, with/without electrical stimulation, providing an ideal matrix for cell growth, proliferation, and tissue deposition. Additionally, these conductive coatings can be enriched with bioactive or pharmaceutical components to enhance the scaffold's biomedical performance. This review covers recent developments in electrically active biomedical coatings for TE. The physicochemical and biological properties of conductive coating materials, including polymers (polypyrrole, polyaniline and PEDOT:PSS), metallic nanoparticles (gold, silver) and inorganic (ceramic) particles (carbon nanotubes, graphene-based materials and Mxenes) are examined. Each section explores the conductive coatings' deposition techniques, deposition parameters, conductivity ranges, deposit morphology, cell responses, and toxicity levels in detail. Furthermore, the applications of these conductive layers, primarily in bone, muscle, and neural TE are considered, and findings from in vitro and in vivo investigations are presented. STATEMENT OF SIGNIFICANCE: Tissue engineering (TE) scaffolds are crucial for human tissue replacement and acceleration of healing. Neural, muscle, bone, and skin tissues have electrically excitable cells, and their regeneration can be enhanced by electrically conductive scaffolds. However, standalone conductive materials often fall short for TE applications. An effective approach involves coating scaffolds with a conductive layer, finely tuning surface properties while leveraging the scaffold's innate biological and physical support. Further enhancement is achieved by modifying the conductive layer with pharmaceutical components. This review explores the under-reviewed topic of conductive coatings in tissue engineering, introducing conductive biomaterial coatings and analyzing their biological interactions. It provides insights into enhancing scaffold functionality for tissue regeneration, bridging a critical gap in current literature.
Collapse
Affiliation(s)
- Abolfazl Anvari Kohestani
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran 11155-4563 Tehran, Iran
| | - Zhiyan Xu
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany
| | - Fatih Erdem Baştan
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany; Thermal Spray Research and Development Laboratory, Metallurgical and Materials Engineering Department, Sakarya University, Esentepe Campus, 54187, Turkey
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen 91058, Germany.
| | - Fatemehsadat Pishbin
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran 11155-4563 Tehran, Iran.
| |
Collapse
|
5
|
Kong C, Guo Z, Teng T, Yao Q, Yu J, Wang M, Ma Y, Wang P, Tang Q. Electroactive Nanomaterials for the Prevention and Treatment of Heart Failure: From Materials and Mechanisms to Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406206. [PMID: 39268781 DOI: 10.1002/smll.202406206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Heart failure (HF) represents a cardiovascular disease that significantly threatens global well-being and quality of life. Electroactive nanomaterials, characterized by their distinctive physical and chemical properties, emerge as promising candidates for HF prevention and management. This review comprehensively examines electroactive nanomaterials and their applications in HF intervention. It presents the definition, classification, and intrinsic characteristics of conductive, piezoelectric, and triboelectric nanomaterials, emphasizing their mechanical robustness, electrical conductivity, and piezoelectric coefficients. The review elucidates their applications and mechanisms: 1) early detection and diagnosis, employing nanomaterial-based sensors for real-time cardiac health monitoring; 2) cardiac tissue repair and regeneration, providing mechanical, chemical, and electrical stimuli for tissue restoration; 3) localized administration of bioactive biomolecules, genes, or pharmacotherapeutic agents, using nanomaterials as advanced drug delivery systems; and 4) electrical stimulation therapies, leveraging their properties for innovative pacemaker and neurostimulation technologies. Challenges in clinical translation, such as biocompatibility, stability, and scalability, are discussed, along with future prospects and potential innovations, including multifunctional and stimuli-responsive nanomaterials for precise HF therapies. This review encapsulates current research and future directions concerning the use of electroactive nanomaterials in HF prevention and management, highlighting their potential to innovating in cardiovascular medicine.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Jiabin Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| |
Collapse
|
6
|
Li YM, Ji Y, Meng YX, Kim YJ, Lee H, Kurian AG, Park JH, Yoon JY, Knowles JC, Choi Y, Kim YS, Yoon BE, Singh RK, Lee HH, Kim HW, Lee JH. Neural Tissue-Like, not Supraphysiological, Electrical Conductivity Stimulates Neuronal Lineage Specification through Calcium Signaling and Epigenetic Modification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400586. [PMID: 38984490 PMCID: PMC11425260 DOI: 10.1002/advs.202400586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Electrical conductivity is a pivotal biophysical factor for neural interfaces, though optimal values remain controversial due to challenges isolating this cue. To address this issue, conductive substrates made of carbon nanotubes and graphene oxide nanoribbons, exhibiting a spectrum of conductivities from 0.02 to 3.2 S m-1, while controlling other surface properties is designed. The focus is to ascertain whether varying conductivity in isolation has any discernable impact on neural lineage specification. Remarkably, neural-tissue-like low conductivity (0.02-0.1 S m-1) prompted neural stem/progenitor cells to exhibit a greater propensity toward neuronal lineage specification (neurons and oligodendrocytes, not astrocytes) compared to high supraphysiological conductivity (3.2 S m-1). High conductivity instigated the apoptotic process, characterized by increased apoptotic fraction and decreased neurogenic morphological features, primarily due to calcium overload. Conversely, cells exposed to physiological conductivity displayed epigenetic changes, specifically increased chromatin openness with H3acetylation (H3ac) and neurogenic-transcription-factor activation, along with a more balanced intracellular calcium response. The pharmacological inhibition of H3ac further supported the idea that such epigenetic changes might play a key role in driving neuronal specification in response to neural-tissue-like, not supraphysiological, conductive cues. These findings underscore the necessity of optimal conductivity when designing neural interfaces and scaffolds to stimulate neuronal differentiation and facilitate the repair process.
Collapse
Affiliation(s)
- Yu-Meng Li
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Yunseong Ji
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Fuel Cell Laboratory, Korea Institute of Energy Research (KIER), Daejeon, 34129, Republic of Korea
| | - Yu-Xuan Meng
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Yu-Jin Kim
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hwalim Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Ji-Young Yoon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Jonathan C Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Hospital, Rowland Hill Street, London, NW3 2PF, UK
| | - Yunkyu Choi
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon-Sik Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Bo-Eun Yoon
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
7
|
Senanayake J, Mattingly RR, Sundararaghavan HG. Electrical stimulation of Schwann cells on electrospun hyaluronic acid carbon nanotube fibers. PLoS One 2024; 19:e0308207. [PMID: 39110684 PMCID: PMC11305570 DOI: 10.1371/journal.pone.0308207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
Neurofibromatosis Type 1 (NF1) is a complex genetic disorder characterized by the development of benign neurofibromas, which can cause significant morbidity in affected individuals. While the molecular mechanisms underlying NF1 pathogenesis have been extensively studied, the development of effective therapeutic strategies remains a challenge. This paper presents the development and validation of a novel biomaterial testing model to enhance our understanding of NF1 pathophysiology, disease mechanisms and evaluate potential therapeutic interventions. Our long-term goal is to develop an invitro model of NF1 to evaluate drug targets. We have developed an in vitro system to test the cellular behavior of NF1 patient derived cells on electroconductive aligned nanofibrous biomaterials with electrical stimulatory cues. We hypothesized that cells cultured on electroconductive biomaterial will undergo morphological changes and variations in cell proliferation that could be further enhanced with the combination of exogenous electrical stimulation (ES). In this study, we developed electrospun Hyaluronic Acid-Carbon Nanotube (HA-CNT) nanofiber scaffolds to mimic the axon's topographical and bioelectrical cues that influence neurofibroma growth and development. The cellular behavior was qualitatively and quantitively analyzed through immunofluorescent stains, Alamar blue assays and ELISA assays. Schwann cells from NF1 patients appear to have lost their ability to respond to electrical stimulation in the development and regeneration range, which was seen through changes in morphology, proliferation and NGF release. Without stimulation, the conductive material enhances NF1 SC behavior. Wild-type SC respond to electrical stimulation with increased cell proliferation and NGF release. Using this system, we can better understand the interaction between axons and SC that lead to tumor formation, homeostasis and regeneration.
Collapse
Affiliation(s)
- Judy Senanayake
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States of America
| | - Raymond R. Mattingly
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, United States of America
| | | |
Collapse
|
8
|
Wang L, Zhao H, Han M, Yang H, Lei M, Wang W, Li K, Li Y, Sang Y, Xin T, Liu H, Qiu J. Electromagnetic Cellularized Patch with Wirelessly Electrical Stimulation for Promoting Neuronal Differentiation and Spinal Cord Injury Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307527. [PMID: 38868910 PMCID: PMC11321663 DOI: 10.1002/advs.202307527] [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: 10/09/2023] [Revised: 04/02/2024] [Indexed: 06/14/2024]
Abstract
Although stem cell therapy holds promise for the treatment of spinal cord injury (SCI), its practical applications are limited by the low degree of neural differentiation. Electrical stimulation is one of the most effective ways to promote the differentiation of stem cells into neurons, but conventional wired electrical stimulation may cause secondary injuries, inflammation, pain, and infection. Here, based on the high conductivity of graphite and the electromagnetic induction effect, graphite nanosheets with neural stem cells (NSCs) are proposed as an electromagnetic cellularized patch to generate in situ wirelessly pulsed electric signals under a rotating magnetic field for regulating neuronal differentiation of NSCs to treat SCI. The strength and frequency of the induced voltage can be controlled by adjusting the rotation speed of the magnetic field. The generated pulsed electrical signals promote the differentiation of NSCs into functional mature neurons and increase the proportion of neurons from 12.5% to 33.7%. When implanted in the subarachnoid region of the injured spinal cord, the electromagnetic cellularized patch improves the behavioral performance of the hind limbs and the repair of spinal cord tissue in SCI mice. This work opens a new avenue for remote treatment of SCI and other nervous system diseases.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Hongbo Zhao
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinan250014P. R. China
| | - Min Han
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinan250014P. R. China
| | - Hongru Yang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Ming Lei
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Wenhan Wang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Keyi Li
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Yiwei Li
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Yuanhua Sang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Tao Xin
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinan250014P. R. China
- Department of Neurosurgery, Shandong Provincial Qianfoshan HospitalShandong UniversityJinan250014P. R. China
- Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinan250117P. R. China
- Department of NeurosurgeryJiangxi Provincial People's HospitalNanchangJiangxi330006P. R. China
| | - Hong Liu
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
- Institute for Advanced Interdisciplinary ResearchUniversity of JinanJinanShandong250022P. R. China
| | - Jichuan Qiu
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| |
Collapse
|
9
|
Wu Y, Zou J, Tang K, Xia Y, Wang X, Song L, Wang J, Wang K, Wang Z. From electricity to vitality: the emerging use of piezoelectric materials in tissue regeneration. BURNS & TRAUMA 2024; 12:tkae013. [PMID: 38957661 PMCID: PMC11218788 DOI: 10.1093/burnst/tkae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 07/04/2024]
Abstract
The unique ability of piezoelectric materials to generate electricity spontaneously has attracted widespread interest in the medical field. In addition to the ability to convert mechanical stress into electrical energy, piezoelectric materials offer the advantages of high sensitivity, stability, accuracy and low power consumption. Because of these characteristics, they are widely applied in devices such as sensors, controllers and actuators. However, piezoelectric materials also show great potential for the medical manufacturing of artificial organs and for tissue regeneration and repair applications. For example, the use of piezoelectric materials in cochlear implants, cardiac pacemakers and other equipment may help to restore body function. Moreover, recent studies have shown that electrical signals play key roles in promoting tissue regeneration. In this context, the application of electrical signals generated by piezoelectric materials in processes such as bone healing, nerve regeneration and skin repair has become a prospective strategy. By mimicking the natural bioelectrical environment, piezoelectric materials can stimulate cell proliferation, differentiation and connection, thereby accelerating the process of self-repair in the body. However, many challenges remain to be overcome before these concepts can be applied in clinical practice, including material selection, biocompatibility and equipment design. On the basis of the principle of electrical signal regulation, this article reviews the definition, mechanism of action, classification, preparation and current biomedical applications of piezoelectric materials and discusses opportunities and challenges for their future clinical translation.
Collapse
Affiliation(s)
- Yifan Wu
- College of Life Sciences, Tiangong University, Binshuixi Road, Xiqing District, Tianjin 300387, China
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Weijin Road, Nankai District, Tianjin 300071, China
| | - Junwu Zou
- College of Life Sciences, Tiangong University, Binshuixi Road, Xiqing District, Tianjin 300387, China
| | - Kai Tang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Fuwai Hospital, Beilishi Road, Xicheng District, Beijing 100037, China
| | - Ying Xia
- College of Life Sciences, Tiangong University, Binshuixi Road, Xiqing District, Tianjin 300387, China
| | - Xixi Wang
- College of Life Sciences, Tiangong University, Binshuixi Road, Xiqing District, Tianjin 300387, China
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Baidi Road, Nankai District, Tianjin 300192, China
| | - Lili Song
- College of Life Sciences, Tiangong University, Binshuixi Road, Xiqing District, Tianjin 300387, China
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Baidi Road, Nankai District, Tianjin 300192, China
| | - Jinhai Wang
- College of Life Sciences, Tiangong University, Binshuixi Road, Xiqing District, Tianjin 300387, China
| | - Kai Wang
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Weijin Road, Nankai District, Tianjin 300071, China
| | - Zhihong Wang
- Institute of Transplant Medicine, School of Medicine, Nankai University, Weijin Road, Nankai District, Tianjin 300071, China
| |
Collapse
|
10
|
Hu C, Liu B, Huang X, Wang Z, Qin K, Sun L, Fan Z. Sea Cucumber-Inspired Microneedle Nerve Guidance Conduit for Synergistically Inhibiting Muscle Atrophy and Promoting Nerve Regeneration. ACS NANO 2024; 18:14427-14440. [PMID: 38776414 DOI: 10.1021/acsnano.4c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Muscle atrophy resulting from peripheral nerve injury (PNI) poses a threat to a patient's mobility and sensitivity. However, an effective method to inhibit muscle atrophy following PNI remains elusive. Drawing inspiration from the sea cucumber, we have integrated microneedles (MNs) and microchannel technology into nerve guidance conduits (NGCs) to develop bionic microneedle NGCs (MNGCs) that emulate the structure and piezoelectric function of sea cucumbers. Morphologically, MNGCs feature an outer surface with outward-pointing needle tips capable of applying electrical stimulation to denervated muscles. Simultaneously, the interior contains microchannels designed to guide the migration of Schwann cells (SCs). Physiologically, the incorporation of conductive reduced graphene oxide and piezoelectric zinc oxide nanoparticles into the polycaprolactone scaffold enhances conductivity and piezoelectric properties, facilitating SCs' migration, myelin regeneration, axon growth, and the restoration of neuromuscular function. These combined effects ultimately lead to the inhibition of muscle atrophy and the restoration of nerve function. Consequently, the concept of the synergistic effect of inhibiting muscle atrophy and promoting nerve regeneration has the capacity to transform the traditional approach to PNI repair and find broad applications in PNI repair.
Collapse
Affiliation(s)
- Cewen Hu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xinyue Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Zhilong Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Kaiqi Qin
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Luyi Sun
- Polymer Program, Institute of Materials Science and Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China
| |
Collapse
|
11
|
Sun J, He L, An Q, Ye X, Ma J, Yan J, Xie X, Sun X, Niu Y, Cao W. Graphene/ chitosan tubes inoculated with dental pulp stem cells promotes repair of facial nerve injury. Front Chem 2024; 12:1417763. [PMID: 38887698 PMCID: PMC11180760 DOI: 10.3389/fchem.2024.1417763] [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: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Introduction: Facial nerve injury significantly impacts both the physical and psychological] wellbeing of patients. Despite advancements, there are still limitations associated with autografts transplantation. Consequently, there is an urgent need for effective artificial grafts to address these limitations and repair injuries. Recent years have witnessed the recognition of the beneficial effects of chitosan (CS) and graphene in the realm of nerve repair. Dental pulp stem cells (DPSCs) hold great promise due to their high proliferative and multi-directional differentiation capabilities. Methods: In this study, Graphene/CS (G/CST) composite tubes were synthesized and their physical, chemical and biological properties were evaluated, then DPSCs were employed as seed cells and G/CST as a scaffold to investigate their combined effect on promoting facial nerve injury repair. Results and Disscussion: The experimental results indicate that G/CST possesses favorable physical and chemical properties, along with good cyto-compatibility. making it suitable for repairing facial nerve transection injuries. Furthermore, the synergistic application of G/CST and DPSCs significantly enhanced the repair process for a 10 mm facial nerve defect in rabbits, highlighting the efficacy of graphene as a reinforcement material and DPSCs as a functional material in facial nerve injury repair. This approach offers an effective treatment strategy and introduces a novel concept for clinically managing facial nerve injuries.
Collapse
Affiliation(s)
- Jingxuan Sun
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Lina He
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Qi An
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xu Ye
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Jinjie Ma
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Jing Yan
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xiaoqi Xie
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Xiangyu Sun
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Yumei Niu
- The First Affiliated Hospital of Harbin Medical University, School of Stomatology, Harbin Medical University, Harbin, China
| | - Wenxin Cao
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology, Harbin, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou, China
| |
Collapse
|
12
|
Kim NY, Choi YY, Kim TH, Ha JH, Kim TH, Kang T, Chung BG. Synergistic Effect of Electrical and Biochemical Stimulation on Human iPSC-Derived Neural Differentiation in a Microfluidic Electrode Array Chip. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15730-15740. [PMID: 38527279 DOI: 10.1021/acsami.3c17108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Neural differentiation is crucial for advancing our understanding of the nervous system and developing treatments for neurological disorders. The advanced methods and the ability to manipulate the alignment, proliferation, and differentiation of stem cells are essential for studying neuronal development and synaptic interactions. However, the utilization of human induced pluripotent stem cells (iPSCs) for disease modeling of neurodegenerative conditions may be constrained by the prolonged duration and uncontrolled cell differentiation required for functional neural cell differentiation. Here, we developed a microfluidic chip to enhance the differentiation and maturation of specific neural lineages by placing aligned microelectrodes on the glass surface to regulate the neural differentiation of human iPSCs. The utilization of electrical stimulation (ES) in conjunction with neurotrophic factors (NF) significantly enhanced the efficiency in generating functional neurons from human iPSCs. We also observed that the simultaneous application of NF and ES to human iPSCs promoted their differentiation and maturation into functional neurons while increasing synaptic interactions. Our research demonstrated the effect of combining NF and ES on human iPSC-derived neural differentiation.
Collapse
Affiliation(s)
- Na Yeon Kim
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| | - Tae Hyeon Kim
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
| | - Jang Ho Ha
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea
| | - Taewook Kang
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea
| | - Bong Geun Chung
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
- Institute of Smart Biosensor, Sogang University, Seoul 04107, Korea
| |
Collapse
|
13
|
De Vitis E, Stanzione A, Romano A, Quattrini A, Gigli G, Moroni L, Gervaso F, Polini A. The Evolution of Technology-Driven In Vitro Models for Neurodegenerative Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304989. [PMID: 38366798 DOI: 10.1002/advs.202304989] [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: 07/21/2023] [Revised: 01/15/2024] [Indexed: 02/18/2024]
Abstract
The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
Collapse
Affiliation(s)
- Eleonora De Vitis
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Antonella Stanzione
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Angelo Quattrini
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Giuseppe Gigli
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Lorenzo Moroni
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, Netherlands
| | - Francesca Gervaso
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
14
|
Shlapakova LE, Surmeneva MA, Kholkin AL, Surmenev RA. Revealing an important role of piezoelectric polymers in nervous-tissue regeneration: A review. Mater Today Bio 2024; 25:100950. [PMID: 38318479 PMCID: PMC10840125 DOI: 10.1016/j.mtbio.2024.100950] [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: 10/05/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Nerve injuries pose a drastic threat to nerve mobility and sensitivity and lead to permanent dysfunction due to low regenerative capacity of mature neurons. The electrical stimuli that can be provided by electroactive materials are some of the most effective tools for the formation of soft tissues, including nerves. Electric output can provide a distinctly favorable bioelectrical microenvironment, which is especially relevant for the nervous system. Piezoelectric biomaterials have attracted attention in the field of neural tissue engineering owing to their biocompatibility and ability to generate piezoelectric surface charges. In this review, an outlook of the most recent achievements in the field of piezoelectric biomaterials is described with an emphasis on piezoelectric polymers for neural tissue engineering. First, general recommendations for the design of an optimal nerve scaffold are discussed. Then, specific mechanisms determining nerve regeneration via piezoelectric stimulation are considered. Activation of piezoelectric responses via natural body movements, ultrasound, and magnetic fillers is also examined. The use of magnetoelectric materials in combination with alternating magnetic fields is thought to be the most promising due to controllable reproducible cyclic deformations and deep tissue permeation by magnetic fields without tissue heating. In vitro and in vivo applications of nerve guidance scaffolds and conduits made of various piezopolymers are reviewed too. Finally, challenges and prospective research directions regarding piezoelectric biomaterials promoting nerve regeneration are discussed. Thus, the most relevant scientific findings and strategies in neural tissue engineering are described here, and this review may serve as a guideline both for researchers and clinicians.
Collapse
Affiliation(s)
- Lada E. Shlapakova
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
| | - Maria A. Surmeneva
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| | - Andrei L. Kholkin
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
- Department of Physics & CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Roman A. Surmenev
- Physical Materials Science and Composite Materials Center, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, Tomsk, 634050, Russia
- Piezo- and Magnetoelectric Materials Research & Development Centre, Research School of Chemistry & Applied Biomedical Sciences, National Research Tomsk Polytechnic University, 634050, Tomsk, Russia
| |
Collapse
|
15
|
Yang S, Zhong S, Jin X, Fan G, Liao X, Yang X, He S. Mapping the hotspots and future trends of electrical stimulation for peripheral nerve injury: A bibliometric analysis from 2002 to 2023. Int Wound J 2024; 21:e14511. [PMID: 38084069 PMCID: PMC10958100 DOI: 10.1111/iwj.14511] [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: 10/06/2023] [Accepted: 11/09/2023] [Indexed: 01/14/2024] Open
Abstract
Peripheral nerve injuries often result in severe personal and social burden, and even with surgical treatment, patients continue to have poor clinical outcomes. Over the past two decades, electrical stimulation has been shown to promote axonal regeneration and alleviate refractory neuropathic pain. The aim of this study was to analyse this field using a bibliometric approach. Literature was searched through Web of Science Core Collection (WOSCC) for the years 2002-2023. Literature analysis included: (1) Describing publication trends in the field. (2) Exploring collaborative network relationships. (3) Finding research advances and research hotspots in the field. (4) Summarizing research trends in the field. With the number of studies in this field still increasing, a total of 693 publications were included in the analysis. This field of research is interdisciplinary in nature. Research hotspots include peripheral nerve regeneration, the treatment of neuropathic pain, materials for nerve injury repair, and the restoration of sensory function in patients with peripheral nerve injury. Correspondingly, the development of nerve conduits and systems for peripheral nerve electrical stimulation, clinical trials of peripheral nerve electrical stimulation, and tactile recovery and movement for amputees have shown significant promise as future research trends in this field.
Collapse
Affiliation(s)
- Sheng Yang
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Sen Zhong
- Shanghai Tongji HospitalTongji University School of MedicineShanghaiChina
| | - Xuehan Jin
- Department of Orthopedic, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Guoxin Fan
- National Key Clinical Pain Medicine of ChinaHuazhong University of Science and Technology Union Shenzhen HospitalShenzhenChina
| | - Xiang Liao
- National Key Clinical Pain Medicine of ChinaHuazhong University of Science and Technology Union Shenzhen HospitalShenzhenChina
| | - Xun Yang
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen Translational Medicine InstituteShenzhen UniversityShenzhenChina
| | - Shisheng He
- Department of Orthopedic, Spinal Pain Research Institute, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
16
|
Zhao Y, Liu Y, Lu C, Sun D, Kang S, Wang X, Lu L. Reduced Graphene Oxide Fibers Combined with Electrical Stimulation Promote Peripheral Nerve Regeneration. Int J Nanomedicine 2024; 19:2341-2357. [PMID: 38469057 PMCID: PMC10926921 DOI: 10.2147/ijn.s449160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/29/2024] [Indexed: 03/13/2024] Open
Abstract
Background The treatment of long-gap peripheral nerve injury (PNI) is still a substantial clinical problem. Graphene-based scaffolds possess extracellular matrix (ECM) characteristic and can conduct electrical signals, therefore have been investigated for repairing PNI. Combined with electrical stimulation (ES), a well performance should be expected. We aimed to determine the effects of reduced graphene oxide fibers (rGOFs) combined with ES on PNI repair in vivo. Methods rGOFs were prepared by one-step dimensionally confined hydrothermal strategy (DCH). Surface characteristics, chemical compositions, electrical and mechanical properties of the samples were characterized. The biocompatibility of the rGOFs were systematically explored both in vitro and in vivo. Total of 54 Sprague-Dawley (SD) rats were randomized into 6 experimental groups: a silicone conduit (S), S+ES, S+rGOFs-filled conduit (SGC), SGC+ES, nerve autograft, and sham groups for a 10-mm sciatic defect. Functional and histological recovery of the regenerated sciatic nerve at 12 weeks after surgery in each group of SD rats were evaluated. Results rGOFs exhibited aligned micro- and nano-channels with excellent mechanical and electrical properties. They are biocompatible in vitro and in vivo. All 6 groups exhibited PNI repair outcomes in view of neurological and morphological recovery. The SGC+ES group achieved similar therapeutic effects as nerve autograft group (P > 0.05), significantly outperformed other treatment groups. Immunohistochemical analysis showed that the expression of proteins related to axonal regeneration and angiogenesis were relatively higher in the SGC+ES. Conclusion The rGOFs had good biocompatibility combined with excellent electrical and mechanical properties. Combined with ES, the rGOFs provided superior motor nerve recovery for a 10-mm nerve gap in a murine acute transection injury model, indicating its excellent repairing ability. That the similar therapeutic effects as autologous nerve transplantation make us believe this method is a promising way to treat peripheral nerve defects, which is expected to guide clinical practice in the future.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yang Liu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Cheng Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Daokuan Sun
- School of Materials Science and Engineering, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Shiqi Kang
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xin Wang
- School of Materials Science and Engineering, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Laijin Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
17
|
Qi W, Zhang R, Wang Z, Du H, Zhao Y, Shi B, Wang Y, Wang X, Wang P. Advances in the Application of Black Phosphorus-Based Composite Biomedical Materials in the Field of Tissue Engineering. Pharmaceuticals (Basel) 2024; 17:242. [PMID: 38399457 PMCID: PMC10892510 DOI: 10.3390/ph17020242] [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: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Black Phosphorus (BP) is a new semiconductor material with excellent biocompatibility, degradability, and optical and electrophysical properties. A growing number of studies show that BP has high potential applications in the biomedical field. This article aims to systematically review the research progress of BP composite medical materials in the field of tissue engineering, mining BP in bone regeneration, skin repair, nerve repair, inflammation, treatment methods, and the application mechanism. Furthermore, the paper discusses the shortcomings and future recommendations related to the development of BP. These shortcomings include stability, photothermal conversion capacity, preparation process, and other related issues. However, despite these challenges, the utilization of BP-based medical materials holds immense promise in revolutionizing the field of tissue repair.
Collapse
Affiliation(s)
- Wanying Qi
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (W.Q.); (R.Z.)
| | - Ru Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (W.Q.); (R.Z.)
| | - Zaishang Wang
- School of Pharmacy, Guilin Medical University, Guilin 541001, China;
| | - Haitao Du
- Shandong Academy of Chinese Medicine, Jinan 250014, China; (H.D.); (Y.Z.); (Y.W.)
| | - Yiwu Zhao
- Shandong Academy of Chinese Medicine, Jinan 250014, China; (H.D.); (Y.Z.); (Y.W.)
| | - Bin Shi
- Shandong Medicinal Biotechnology Center, Jinan 250062, China;
| | - Yi Wang
- Shandong Academy of Chinese Medicine, Jinan 250014, China; (H.D.); (Y.Z.); (Y.W.)
| | - Xin Wang
- State Key Laboratory of Biobased Material and Green Papermaking, Faculty of Light Industry, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Ping Wang
- Shandong Academy of Chinese Medicine, Jinan 250014, China; (H.D.); (Y.Z.); (Y.W.)
| |
Collapse
|
18
|
Wang Y, Yang B, Huang Z, Yang Z, Wang J, Ao Q, Yin G, Li Y. Progress and mechanism of graphene oxide-composited materials in application of peripheral nerve repair. Colloids Surf B Biointerfaces 2024; 234:113672. [PMID: 38071946 DOI: 10.1016/j.colsurfb.2023.113672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/09/2024]
Abstract
Peripheral nerve injuries (PNI) are one of the most common nerve injuries, and graphene oxide (GO) has demonstrated significant potential in the treatment of PNI. GO could enhance the proliferation, adhesion, migration, and differentiation of neuronal cells by upregulating the expression of relevant proteins, and regulate the angiogenesis process and immune response. Therefore, GO is a suitable additional component for fabricating artificial nerve scaffolds (ANS), in which the slight addition of GO could improve the physicochemical performance of the matrix materials, through hydrogen bonds and electrostatic attraction. GO-composited ANS can increase the expression of nerve regeneration-associated genes and factors, promoting angiogenesis by activating the RAS/MAPK and AKT-eNOS-VEGF signaling pathway, respectively. Moreover, GO could be metabolized and excreted from the body through the pathway of peroxidase degradation in vivo. Consequently, the application of GO in PNI regeneration exhibits significant potential for transitioning from laboratory research to clinical use.
Collapse
Affiliation(s)
- Yulin Wang
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| | - Bing Yang
- College of Biomedical Engineering, Sichuan University, China; Precision Medical Center of Southwest China Hospital, Sichuan University, China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, China.
| | - Zhaopu Yang
- Center for Drug Inspection, Guizhou Medical Products Administration, China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, China
| | - Qiang Ao
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, China
| | - Ya Li
- College of Biomedical Engineering, Sichuan University, China; Institute of Regulatory Science for Medical Devices, Sichuan University, China
| |
Collapse
|
19
|
Deleye L, Franchi F, Trevisani M, Loiacono F, Vercellino S, Debellis D, Liessi N, Armirotti A, Vázquez E, Valente P, Castagnola V, Benfenati F. Few-layered graphene increases the response of nociceptive neurons to irritant stimuli. NANOSCALE 2024; 16:2419-2431. [PMID: 38226500 DOI: 10.1039/d3nr03790h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The unique properties of few-layered graphene (FLG) make it interesting for a variety of applications, including biomedical applications, such as tissue engineering and drug delivery. Although different studies focus on applications in the central nervous system, its interaction with the peripheral nervous system has been so far overlooked. Here, we investigated the effects of exposure to colloidal dispersions of FLG on the sensory neurons of the rat dorsal root ganglia (DRG). We found that the FLG flakes were actively internalized by sensory neurons, accumulated in large intracellular vesicles, and possibly degraded over time, without major toxicological concerns, as neuronal viability, morphology, protein content, and basic electrical properties of DRG neurons were preserved. Interestingly, in our electrophysiological investigation under noxious stimuli, we observed an increased functional response upon FLG treatment of the nociceptive subpopulation of DRG neurons in response to irritants specific for chemoreceptors TRPV1 and TRPA1. The observed effects of FLG on DRG neurons may open-up novel opportunities for applications of these materials in specific disease models.
Collapse
Affiliation(s)
- Lieselot Deleye
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
| | - Francesca Franchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Martina Trevisani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genoa, 16132, Italy.
| | - Fabrizio Loiacono
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Silvia Vercellino
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, IIT, Via Morego 30, 16163, Genoa, Italy
| | - Nara Liessi
- Analytical Chemistry Facility, IIT, via Morego, 30, 16163, Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, IIT, via Morego, 30, 16163, Genoa, Italy
| | - Ester Vázquez
- Facultad de Ciencias Químicas, Universidad Castilla La-Mancha, Ciudad Real, 13071 Spain
| | - Pierluigi Valente
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genoa, 16132, Italy.
| | - Valentina Castagnola
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
20
|
Convertino D, Nencioni M, Russo L, Mishra N, Hiltunen VM, Bertilacchi MS, Marchetti L, Giacomelli C, Trincavelli ML, Coletti C. Interaction of graphene and WS 2 with neutrophils and mesenchymal stem cells: implications for peripheral nerve regeneration. NANOSCALE 2024; 16:1792-1806. [PMID: 38175567 DOI: 10.1039/d3nr04927b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Graphene and bidimensional (2D) materials have been widely used in nerve conduits to boost peripheral nerve regeneration. Nevertheless, the experimental and commercial variability in graphene-based materials generates graphene forms with different structures and properties that can trigger entirely diverse biological responses from all the players involved in nerve repair. Herein, we focus on the graphene and tungsten disulfide (WS2) interaction with non-neuronal cell types involved in nerve tissue regeneration. We synthesize highly crystalline graphene and WS2 with scalable techniques such as thermal decomposition and chemical vapor deposition. The materials were able to trigger the activation of a neutrophil human model promoting Neutrophil Extracellular Traps (NETs) production, particularly under basal conditions, although neutrophils were not able to degrade graphene. Of note is that pristine graphene acts as a repellent for the NET adhesion, a beneficial property for nerve conduit long-term applications. Mesenchymal stem cells (MSCs) have been proposed as a promising strategy for nerve regeneration in combination with a conduit. Thus, the interaction of graphene with MSCs was also investigated, and reduced viability was observed only on specific graphene substrates. Overall, the results confirm the possibility of regulating the cell response by varying graphene properties and selecting the most suitable graphene forms.
Collapse
Affiliation(s)
- Domenica Convertino
- Center for Nanotechnology Innovation @ NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy.
| | - Martina Nencioni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy.
| | - Lara Russo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy.
| | - Neeraj Mishra
- Center for Nanotechnology Innovation @ NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy.
- Graphene Labs, Istituto Italiano di Tecnologia, Via Morego 30, Genova, Italy
| | - Vesa-Matti Hiltunen
- Center for Nanotechnology Innovation @ NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy.
- Graphene Labs, Istituto Italiano di Tecnologia, Via Morego 30, Genova, Italy
| | | | - Laura Marchetti
- Center for Nanotechnology Innovation @ NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy.
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy.
| | - Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa, Italy.
| | | | - Camilla Coletti
- Center for Nanotechnology Innovation @ NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, Pisa, Italy.
- Graphene Labs, Istituto Italiano di Tecnologia, Via Morego 30, Genova, Italy
| |
Collapse
|
21
|
Song Y, Liu L, Li S, Jiang X, Zheng X. CoFeSe 2 @DMSA@FA Nanocatalyst for Amplification of Oxidative Stress to Achieve Multimodal Tumor Therapy. Chembiochem 2024; 25:e202300631. [PMID: 37930640 DOI: 10.1002/cbic.202300631] [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: 09/15/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 11/07/2023]
Abstract
Nanomedicine has significantly advanced precise tumor therapy, providing essential technical blessing for active drug accumulation, targeted consignment, and mitigation of noxious side effects. To enhance anti-tumor efficacy, the integration of multiple therapeutic modalities has garnered significant attention. Here, we designed an innovative CoFeSe2 @DMSA@FA nanocatalyst with Se vacancies (abbreviated as CFSDF), which exhibits synergistic chemodynamic therapy (CDT) and photothermal therapy (PTT), leading to amplified tumor oxidative stress and enhanced photothermal effects. The multifunctional CFSDF nanocatalyst exhibits the remarkable ability to catalyze the Fenton reaction within the acidic tumor microenvironment, efficiently converting hydrogen peroxide (H2 O2 ) into highly harmful hydroxyl radicals (⋅OH). Moreover, the nanocatalyst effectively diminishes GSH levels and ameliorates intracellular oxidative stress. The incorporation of FA modification enables CFSDF to evade immune detection and selectively target tumor tissues. Numerous in vitro and in vivo investigations have consistently demonstrated that CFSDF optimizes its individual advantages and significantly enhances therapeutic efficiency through synergistic effects of multiple therapeutic modalities, offering a valuable and effective approach to cancer treatment.
Collapse
Affiliation(s)
- Yingzi Song
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276000, China
- Key Laboratory of Advanced Biomaterials and, Nanomedicine in Universities of Shandong, Linyi University, Linyi, 276000, China
| | - Lekang Liu
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276000, China
- Key Laboratory of Advanced Biomaterials and, Nanomedicine in Universities of Shandong, Linyi University, Linyi, 276000, China
| | - Shulian Li
- Linyi Cancer Hospital, Linyi, 276000, China) E-mail: address
| | - Xiaolei Jiang
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276000, China
- Key Laboratory of Advanced Biomaterials and, Nanomedicine in Universities of Shandong, Linyi University, Linyi, 276000, China
| | - Xiuwen Zheng
- Key Laboratory of Advanced Biomaterials and, Nanomedicine in Universities of Shandong, Linyi University, Linyi, 276000, China
- Qilu Normal University, Jinan, 250200, China
| |
Collapse
|
22
|
Marques-Almeida T, Lanceros-Mendez S, Ribeiro C. State of the Art and Current Challenges on Electroactive Biomaterials and Strategies for Neural Tissue Regeneration. Adv Healthc Mater 2024; 13:e2301494. [PMID: 37843074 DOI: 10.1002/adhm.202301494] [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: 05/09/2023] [Revised: 09/22/2023] [Indexed: 10/17/2023]
Abstract
The loss or failure of an organ/tissue stands as one of the healthcare system's most prevalent, devastating, and costly challenges. Strategies for neural tissue repair and regeneration have received significant attention due to their particularly strong impact on patients' well-being. Many research efforts are dedicated not only to control the disease symptoms but also to find solutions to repair the damaged tissues. Neural tissue engineering (TE) plays a key role in addressing this problem and significant efforts are being carried out to develop strategies for neural repair treatment. In the last years, active materials allowing to tune cell-materials interaction are being increasingly used, representing a recent paradigm in TE applications. Among the most important stimuli influencing cell behavior are the electrical and mechanical ones. In this way, materials with the ability to provide this kind of stimuli to the neural cells seem to be appropriate to support neural TE. In this scope, this review summarizes the different biomaterials types used for neural TE, highlighting the relevance of using active biomaterials and electrical stimulation. Furthermore, this review provides not only a compilation of the most relevant studies and results but also strategies for novel and more biomimetic approaches for neural TE.
Collapse
Affiliation(s)
- Teresa Marques-Almeida
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| | - Senentxu Lanceros-Mendez
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, 48940, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain
| | - Clarisse Ribeiro
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| |
Collapse
|
23
|
Deng W, Li X, Li Y, Huang Z, Wang Y, Mu N, Wang J, Chen T, Pu X, Yin G, Feng H. Graphene oxide-doped chiral dextro-hydrogel promotes peripheral nerve repair through M2 polarization of macrophages. Colloids Surf B Biointerfaces 2024; 233:113632. [PMID: 37979485 DOI: 10.1016/j.colsurfb.2023.113632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/20/2023]
Abstract
Dextro-chirality is reported to specifically promote the proliferation and survival of neural cells. However, applying this unique performance to nerve repair remains a great challenge. Graphite oxide (GO)-phenylalanine derivative hydrogel system was constructed through doping 5% GO into self-assembly dextro- or levo-hydrogels (named as dextro and levo group, respectively), which exhibited identical physical and chemical properties, cyto-compatibility, and mirror-symmetrical chirality. In vivo experiments using rat sciatic nerve repair models showed that the functional recovery and histological restoration of regenerating nerves in the dextro group were significantly improved, approaching that of autograft implantation. The doped GO promoted M2 polarization of macrophages, increasing the expression of platelet-derived growth factor BB chain and vascular endothelial growth factor, thereby improving angiogenesis in regenerating nerves. A mechanism is proposed for the facilitated nerve repair through the synergistic effect of GO and dextro-hydrogel, involving dextro-chirality selection of neural cells and GO-induced M2 polarization, which promotes microvascular regeneration and myelination. This study showcases the immense potential of chirality in addressing neurological issues by providing a compelling demonstration of the development of effective therapies that leverage the unique matrix chirality selection of nerve cells to promote peripheral nerve regeneration.
Collapse
Affiliation(s)
- Weiping Deng
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Xiaohui Li
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Ya Li
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China.
| | - Yulin Wang
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Ning Mu
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China; Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), No. 29, Gaotanyanzheng Street, Shapingba District, Chongqing 400038, China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Tunan Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), No. 29, Gaotanyanzheng Street, Shapingba District, Chongqing 400038, China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, No. 24, South 1st Section, 1st Ring Road, Chengdu 610065, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), No. 29, Gaotanyanzheng Street, Shapingba District, Chongqing 400038, China
| |
Collapse
|
24
|
Kang MS, Jang HJ, Jo HJ, Raja IS, Han DW. MXene and Xene: promising frontier beyond graphene in tissue engineering and regenerative medicine. NANOSCALE HORIZONS 2023; 9:93-117. [PMID: 38032647 DOI: 10.1039/d3nh00428g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
The emergence of 2D nanomaterials (2D NMs), which was initiated by the isolation of graphene (G) in 2004, revolutionized various biomedical applications, including bioimaging and -sensing, drug delivery, and tissue engineering, owing to their unique physicochemical and biological properties. Building on the success of G, a novel class of monoelemental 2D NMs, known as Xenes, has recently emerged, offering distinct advantages in the fields of tissue engineering and regenerative medicine. In this review, we focus on the comparison of G and Xene materials for use in fabricating tissue engineering scaffolds. After a brief introduction to the basic physicochemical properties of these materials, recent representative studies are classified in terms of the engineered tissue, i.e., bone, cartilage, neural, muscle, and skin tissues. We analyze several methods of improving the clinical potential of Xene-laden scaffolds using state-of-the-art fabrication technologies and innovative biomaterials. Despite the considerable advantages of Xene materials, critical concerns, such as biocompatibility, biodistribution and regulatory challenges, should be considered. This review and collaborative efforts should advance the field of Xene-based tissue engineering and enable innovative, effective solutions for use in future tissue regeneration.
Collapse
Affiliation(s)
- Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea.
| | - Hee Jeong Jang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea.
| | - Hyo Jung Jo
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea.
| | | | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea.
- BIO-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
25
|
Convertino D, Trincavelli ML, Giacomelli C, Marchetti L, Coletti C. Graphene-based nanomaterials for peripheral nerve regeneration. Front Bioeng Biotechnol 2023; 11:1306184. [PMID: 38164403 PMCID: PMC10757979 DOI: 10.3389/fbioe.2023.1306184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
Emerging nanotechnologies offer numerous opportunities in the field of regenerative medicine and have been widely explored to design novel scaffolds for the regeneration and stimulation of nerve tissue. In this review, we focus on peripheral nerve regeneration. First, we introduce the biomedical problem and the present status of nerve conduits that can be used to guide, fasten and enhance regeneration. Then, we thoroughly discuss graphene as an emerging candidate in nerve tissue engineering, in light of its chemical, tribological and electrical properties. We introduce the graphene forms commonly used as neural interfaces, briefly review their applications, and discuss their potential toxicity. We then focus on the adoption of graphene in peripheral nervous system applications, a research field that has gained in the last years ever-increasing attention. We discuss the potential integration of graphene in guidance conduits, and critically review graphene interaction not only with peripheral neurons, but also with non-neural cells involved in nerve regeneration; indeed, the latter have recently emerged as central players in modulating the immune and inflammatory response and accelerating the growth of new tissue.
Collapse
Affiliation(s)
- Domenica Convertino
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Pisa, Italy
| | | | | | - Laura Marchetti
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Pisa, Italy
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Camilla Coletti
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Pisa, Italy
| |
Collapse
|
26
|
Rahman M, Mahady Dip T, Padhye R, Houshyar S. Review on electrically conductive smart nerve guide conduit for peripheral nerve regeneration. J Biomed Mater Res A 2023; 111:1916-1950. [PMID: 37555548 DOI: 10.1002/jbm.a.37595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/29/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023]
Abstract
At present, peripheral nerve injuries (PNIs) are one of the leading causes of substantial impairment around the globe. Complete recovery of nerve function after an injury is challenging. Currently, autologous nerve grafts are being used as a treatment; however, this has several downsides, for example, donor site morbidity, shortage of donor sites, loss of sensation, inflammation, and neuroma development. The most promising alternative is the development of a nerve guide conduit (NGC) to direct the restoration and renewal of neuronal axons from the proximal to the distal end to facilitate nerve regeneration and maximize sensory and functional recovery. Alternatively, the response of nerve cells to electrical stimulation (ES) has a substantial regenerative effect. The incorporation of electrically conductive biomaterials in the fabrication of smart NGCs facilitates the function of ES throughout the active proliferation state. This article overviews the potency of the various categories of electroactive smart biomaterials, including conductive and piezoelectric nanomaterials, piezoelectric polymers, and organic conductive polymers that researchers have employed latterly to fabricate smart NGCs and their potentiality in future clinical application. It also summarizes a comprehensive analysis of the recent research and advancements in the application of ES in the field of NGC.
Collapse
Affiliation(s)
- Mustafijur Rahman
- Center for Materials Innovation and Future Fashion (CMIFF), School of Fashion and Textiles, RMIT University, Brunswick, Australia
- Department of Dyes and Chemical Engineering, Bangladesh University of Textiles, Dhaka, Bangladesh
| | - Tanvir Mahady Dip
- Department of Materials, University of Manchester, Manchester, UK
- Department of Yarn Engineering, Bangladesh University of Textiles, Dhaka, Bangladesh
| | - Rajiv Padhye
- Center for Materials Innovation and Future Fashion (CMIFF), School of Fashion and Textiles, RMIT University, Brunswick, Australia
| | - Shadi Houshyar
- School of Engineering, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Eftekhari BS, Song D, Janmey PA. Electrical Stimulation of Human Mesenchymal Stem Cells on Conductive Substrates Promotes Neural Priming. Macromol Biosci 2023; 23:e2300149. [PMID: 37571815 PMCID: PMC10880582 DOI: 10.1002/mabi.202300149] [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: 04/06/2023] [Revised: 06/29/2023] [Indexed: 08/13/2023]
Abstract
Electrical stimulation (ES) within a conductive scaffold is potentially beneficial in encouraging the differentiation of stem cells toward a neuronal phenotype. To improve stem cell-based regenerative therapies, it is essential to use electroconductive scaffolds with appropriate stiffnesses to regulate the amount and location of ES delivery. Herein, biodegradable electroconductive substrates with different stiffnesses are fabricated from chitosan-grafted-polyaniline (CS-g-PANI) copolymers. Human mesenchymal stem cells (hMSCs) cultured on soft conductive scaffolds show a morphological change with significant filopodial elongation after electrically stimulated culture along with upregulation of neuronal markers and downregulation of glial markers. Compared to stiff conductive scaffolds and non-conductive CS scaffolds, soft conductive CS-g-PANI scaffolds promote increased expression of microtubule-associated protein 2 (MAP2) and neurofilament heavy chain (NF-H) after application of ES. At the same time, there is a decrease in the expression of the glial markers glial fibrillary acidic protein (GFAP) and vimentin after ES. Furthermore, the elevation of intracellular calcium [Ca2+ ] during spontaneous, cell-generated Ca2+ transients further suggests that electric field stimulation of hMSCs cultured on conductive substrates can promote a neural-like phenotype. The findings suggest that the combination of the soft conductive CS-g-PANI substrate and ES is a promising new tool for enhancing neuronal tissue engineering outcomes.
Collapse
Affiliation(s)
| | - Dawei Song
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A. Janmey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Shi S, Ou X, Cheng D. How Advancing is Peripheral Nerve Regeneration Using Nanofiber Scaffolds? A Comprehensive Review of the Literature. Int J Nanomedicine 2023; 18:6763-6779. [PMID: 38026517 PMCID: PMC10657550 DOI: 10.2147/ijn.s436871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Peripheral nerve injuries present significant challenges in regenerative medicine, primarily due to inherent limitations in the body's natural healing processes. In response to these challenges and with the aim of enhancing peripheral nerve regeneration, nanofiber scaffolds have emerged as a promising and advanced intervention. However, a deeper understanding of the underlying mechanistic foundations that drive the favorable contributions of nanofiber scaffolds to nerve regeneration is essential. In this comprehensive review, we make an exploration of the latent potential of nanofiber scaffolds in augmenting peripheral nerve regeneration. This exploration includes a detailed introduction to the fabrication methods of nanofibers, an analysis of the intricate interactions between these scaffolds and cellular entities, an examination of strategies related to the controlled release of bioactive agents, an assessment of the prospects for clinical translation, an exploration of emerging trends, and thorough considerations regarding biocompatibility and safety. By comprehensively elucidating the intricate structural attributes and multifaceted functional capacities inherent in nanofiber scaffolds, we aim to offer a prospective and effective strategy for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Shaoyan Shi
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Deliang Cheng
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| |
Collapse
|
29
|
Rosenbalm TN, Levi NH, Morykwas MJ, Wagner WD. Electrical stimulation via repeated biphasic conducting materials for peripheral nerve regeneration. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:61. [PMID: 37964030 PMCID: PMC10645611 DOI: 10.1007/s10856-023-06763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
Improved materials for peripheral nerve repair are needed for the advancement of new surgical techniques in fields spanning from oncology to trauma. In this study, we developed bioresorbable materials capable of producing repeated electric field gradients spaced 600 μm apart to assess the impact on neuronal cell growth, and migration. Electrically conductive, biphasic composites comprised of poly (glycerol) sebacate acrylate (PGSA) alone, and doped with poly (pyrrole) (PPy), were prepared to create alternating segments with high and low electrically conductivity. Conductivity measurements demonstrated that 0.05% PPy added to PSA achieved an optimal value of 1.25 × 10-4 S/cm, for subsequent electrical stimulation. Tensile testing and degradation of PPy doped and undoped PGSA determined that 35-40% acrylation of PGSA matched nerve mechanical properties. Both fibroblast and neuronal cells thrived when cultured upon the composite. Biphasic PGSA/PPy sheets seeded with neuronal cells stimulated for with 3 V, 20 Hz demonstrated a 5x cell increase with 1 day of stimulation and up to a 10x cell increase with 3 days stimulation compared to non-stimulated composites. Tubular conduits composed of repeated high and low conductivity materials suitable for implantation in the rat sciatic nerve model for nerve repair were evaluated in vivo and were superior to silicone conduits. These results suggest that biphasic conducting conduits capable of maintaining mechanical properties without inducing compression injuries while generating repeated electric fields are a promising tool for acceleration of peripheral nerve repair to previously untreatable patients.
Collapse
Affiliation(s)
- Tabitha N Rosenbalm
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Polytechnic Institute and State University, Winston-Salem, NC, 27106, USA
- Department of Plastic and Reconstructive Surgery, Wake Forest Baptist Health, Winston-Salem, NC, 27157, USA
| | - Nicole H Levi
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Polytechnic Institute and State University, Winston-Salem, NC, 27106, USA.
- Department of Plastic and Reconstructive Surgery, Wake Forest Baptist Health, Winston-Salem, NC, 27157, USA.
| | - Michael J Morykwas
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Polytechnic Institute and State University, Winston-Salem, NC, 27106, USA
- Department of Plastic and Reconstructive Surgery, Wake Forest Baptist Health, Winston-Salem, NC, 27157, USA
| | - William D Wagner
- School of Biomedical Engineering and Sciences, Wake Forest University-Virginia Polytechnic Institute and State University, Winston-Salem, NC, 27106, USA
- Department of Plastic and Reconstructive Surgery, Wake Forest Baptist Health, Winston-Salem, NC, 27157, USA
| |
Collapse
|
30
|
Lan D, Wu B, Zhang H, Chen X, Li Z, Dai F. Novel Bioinspired Nerve Scaffold with High Synchrony between Biodegradation and Nerve Regeneration for Repair of Peripheral Nerve Injury. Biomacromolecules 2023; 24:5451-5466. [PMID: 37917398 DOI: 10.1021/acs.biomac.3c00920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
The morphological structure reconstruction and functional recovery of long-distance peripheral nerve injury (PNI) are global medical challenges. Biodegradable nerve scaffolds that provide mechanical support for the growth and extension of neurites are a desired way to repair long-distance PNI. However, the synchrony of scaffold degradation and nerve regeneration is still challenging. Here, a novel bioinspired multichannel nerve guide conduit (MNGC) with topographical cues based on silk fibroin and ε-polylysine modification was constructed. This conduit (SF(A) + PLL MNGC) exhibited sufficient mechanical strength, excellent degradability, and favorable promotion of cell growth. Peripheral nerve repairing was evaluated by an in vivo 10 mm rat sciatic model. In vivo evidence demonstrated that SF(A) + PLL MNGC was completely biodegraded in the body within 4 weeks after providing sufficient physical support and guide for neurite extension, and a 10 mm sciatic nerve defect was effectively repaired without scar formation, indicating a high synchronous effect of scaffold biodegradation and nerve regeneration. More importantly, the regenerated nerve of the SF(A) + PLL MNGC group showed comparable morphological reconstruction and functional recovery to that of autologous nerve transplantation. This work proved that the designed SF(A) + PLL MNGC has potential for application in long-distance PNI repair in the clinic.
Collapse
Affiliation(s)
- Dongwei Lan
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Baiqing Wu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Haiqiang Zhang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Xiang Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Zhi Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Fangyin Dai
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
- Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China
- College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| |
Collapse
|
31
|
Wang J, Wang Z, Li Y, Hou Y, Yin C, Yang E, Liao Z, Fan C, Martin LL, Sun D. Blood brain barrier-targeted delivery of double selenium nanospheres ameliorates neural ferroptosis in Alzheimer's disease. Biomaterials 2023; 302:122359. [PMID: 39491374 DOI: 10.1016/j.biomaterials.2023.122359] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 09/28/2023] [Accepted: 10/17/2023] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) as a common neurodegenerative disease showed progressive cognitive dysfunction and behavioral impairment. Currently, the deposition of amyloid β-protein (Aβ) remains the main pathomechanism. However, preventing neuronal death induced by Aβ remains elusive, and no effective strategy in clinic was found to combat AD. Herein, a multifunctional double selenium nanosphere (CLNDSe) was designed and prepared, and A2AAR agonist (CGS) modification endowed CLNDSe NPs with A2AAR-targeted blood brain barrier (BBB) delivery in vitro and in vivo. CLNDSe NPs after modification of LPFFD short peptide effectively inhibited Aβ42 aggregation and attenuated Aβ42-induced neural toxicity by inhibiting oxidative damage and mitochondrial dysfunctions. Nerve growth factor (NGF) linked to large Se sphere significantly attenuated Tau phosphorylation and gliocytes activation in APP/PS1 mice. CLNDSe NPs administration in vivo also effectively restored GPX1/4 antioxidant ability, alleviated neural loss and neurofibrillary tangles, prevented neural ferroptosis, and eventually ameliorated cognitive deficits of APP/PS1 mice. Importantly, CLNDSe NPs showed good safety and biocompatibility. Taken together, our finding validated the rational design that BBB-targeted delivery of double selenium nanosphere may be a novel strategy to ameliorate Alzheimer's disease by inhibiting neural ferroptosis.
Collapse
Affiliation(s)
- Jingyuan Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Zekun Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Yuqing Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Yajun Hou
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Chenyang Yin
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Endong Yang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Ziyu Liao
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Cundong Fan
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Lisandra L Martin
- School of Chemistry, Monash University, Clayton, 3800, Victoria, Australia.
| | - Dongdong Sun
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China.
| |
Collapse
|
32
|
Mao W, Lee E, Cho W, Kang BJ, Yoo HS. Cell-directed assembly of luminal nanofibril fillers in nerve conduits for peripheral nerve repair. Biomaterials 2023; 301:122209. [PMID: 37421670 DOI: 10.1016/j.biomaterials.2023.122209] [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: 11/24/2022] [Revised: 06/01/2023] [Accepted: 06/16/2023] [Indexed: 07/10/2023]
Abstract
Graphene and its derivatives, graphene oxide (GO) and reduced graphene oxide (rGO), have attracted significant attention in the field of tissue engineering, particularly in nerve and muscle regeneration, owing to their excellent electrical conductivity. This paper reports the fabrication of cell-mixable rGO-decorated polycaprolactone (PCL) nanofibrils (NFs) to promote peripheral nerve repair with the assistant of electron transmission by rGO and cytokine paracrine by stem cells. Oxidized GO (GO-COOH) and branched polyethylenimine are layer-by-layer coated on hydrolyzed PCL NFs via electrostatic interaction, and the number of layering is manipulated to adjust the GO-COOH coating amount. The decorated GO-COOH is reduced in situ to rGO for electrical conductivity retrieval. PC12 cells cultivated with rGO-coated NF demonstrate spontaneous cell sheet assembly, and neurogenic differentiation is observed upon electrical stimulation. When transplant nerve guidance conduit containing the assembly of rGO-coated NF and adipose-derived stem cell to the site of neurotmesis injury of a sciatic nerve, animal movement is enhanced and autotomy is ameliorated for 8 weeks compared to transplanting the hollow conduit only. Histological analysis results reveal higher levels of muscle mass and lower levels of collagen deposition in the triceps surae muscle of the rGO-coated NF-treated legs. Therefore, the rGO-layered NF can be tailored to repair peripheral nerve injuries in combination with stem cell therapy.
Collapse
Affiliation(s)
- Wei Mao
- Department of Biomedical Materials Engineering, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Eunbee Lee
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea; BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wanho Cho
- Department of Biomedical Materials Engineering, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea; BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Hyuk Sang Yoo
- Department of Biomedical Materials Engineering, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea; Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
33
|
Mankavi F, Ibrahim R, Wang H. Advances in Biomimetic Nerve Guidance Conduits for Peripheral Nerve Regeneration. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2528. [PMID: 37764557 PMCID: PMC10536071 DOI: 10.3390/nano13182528] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Injuries to the peripheral nervous system are a common clinical issue, causing dysfunctions of the motor and sensory systems. Surgical interventions such as nerve autografting are necessary to repair damaged nerves. Even with autografting, i.e., the gold standard, malfunctioning and mismatches between the injured and donor nerves often lead to unwanted failure. Thus, there is an urgent need for a new intervention in clinical practice to achieve full functional recovery. Nerve guidance conduits (NGCs), providing physicochemical cues to guide neural regeneration, have great potential for the clinical regeneration of peripheral nerves. Typically, NGCs are tubular structures with various configurations to create a microenvironment that induces the oriented and accelerated growth of axons and promotes neuron cell migration and tissue maturation within the injured tissue. Once the native neural environment is better understood, ideal NGCs should maximally recapitulate those key physiological attributes for better neural regeneration. Indeed, NGC design has evolved from solely physical guidance to biochemical stimulation. NGC fabrication requires fundamental considerations of distinct nerve structures, the associated extracellular compositions (extracellular matrices, growth factors, and cytokines), cellular components, and advanced fabrication technologies that can mimic the structure and morphology of native extracellular matrices. Thus, this review mainly summarizes the recent advances in the state-of-the-art NGCs in terms of biomaterial innovations, structural design, and advanced fabrication technologies and provides an in-depth discussion of cellular responses (adhesion, spreading, and alignment) to such biomimetic cues for neural regeneration and repair.
Collapse
Affiliation(s)
| | | | - Hongjun Wang
- Department of Biomedical Engineering, Semcer Center for Healthcare Innovation, Stevens Institute of Technology, Hoboken, NJ 07030, USA; (F.M.); (R.I.)
| |
Collapse
|
34
|
Kim J, Jeon J, Lee J, Khoroldulam B, Choi S, Bae J, Hyun JK, Kang S. Electroceuticals for Regeneration of Long Nerve Gap Using Biodegradable Conductive Conduits and Implantable Wireless Stimulator. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302632. [PMID: 37340589 PMCID: PMC10460856 DOI: 10.1002/advs.202302632] [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: 04/25/2023] [Revised: 12/12/2012] [Indexed: 06/22/2023]
Abstract
Regeneration of over 10 mm long peripheral nerve defects remains a challenge due to the failure of regeneration by prolonged axotomy and denervation occurring in long-term recovery. Recent studies reveal that conductive conduits and electrical stimulation accelerate the regeneration of long nerve defects. In this study, an electroceutical platform combining a fully biodegradable conductive nerve conduit and a wireless electrical stimulator is proposed to maximize the therapeutic effect on nerve regeneration. Fully biodegradable nerve conduit fabricated using molybdenum (Mo) microparticles and polycaprolactone (PCL) can eliminate the unwanted effects of non-degradable implants, which occupy nerve paths and need to be removed through surgery increasing the risk of complications. The electrical and mechanical properties of Mo/PCL conduits are optimized by controlling the amounts of Mo and tetraglycol lubricant. The dissolution behavior and electrical conductivity of biodegradable nerve conduits in the biomimetic solutions are also evaluated. In in vivo experiments, the integrated strategy of a conductive Mo/PCL conduit with controlled therapeutic electrical stimulation shows accelerated axon regeneration for long sciatic nerve defects in rats compared to the use of the Mo/PCL conduit without stimulation and has a significant therapeutic effect based on the results obtained from the functional recovery test.
Collapse
Affiliation(s)
- Jio Kim
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Jooik Jeon
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Ju‐Yong Lee
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Badamgarav Khoroldulam
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
| | - Sung‐Geun Choi
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Jae‐Young Bae
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Jung Keun Hyun
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative MedicineDankook UniversityCheonan31116Republic of Korea
- Department of Rehabilitation MedicineCollege of MedicineDankook UniversityCheonan31116Republic of Korea
- Institute of Tissue Regeneration Engineering (ITREN)Dankook UniversityCheonan31116Republic of Korea
| | - Seung‐Kyun Kang
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
- Research Institute of Advanced Materials (RIAM)Seoul National UniversitySeoul08826Republic of Korea
- Nano Systems Institute SOFT FoundrySeoul National UniversitySeoul08826Republic of korea
| |
Collapse
|
35
|
Öksüz KE, Kurt B, Şahin İnan ZD, Hepokur C. Novel Bioactive Glass/Graphene Oxide-Coated Surgical Sutures for Soft Tissue Regeneration. ACS OMEGA 2023; 8:21628-21641. [PMID: 37360470 PMCID: PMC10286287 DOI: 10.1021/acsomega.3c00978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023]
Abstract
The combination of a commercially available PGLA (poly[glycolide-co-l-lactide]), 90:10% suture material with bioactive bioglass nanopowders (BGNs) and graphene oxide (GO)-doped BGNs offers new opportunities for the clinical application of biomaterials in soft tissue engineering. In the present experimental work, we demonstrate that GO-doped melt-derived BGNs were synthesized via the sol-gel process. After that, novel GO-doped and undoped BGNs were used to coat resorbable PGLA surgical sutures, thereby imparting bioactivity, biocompatibility, and accelerated wound healing properties to the sutures. Stable and homogeneous coatings on the surface of the sutures were achieved using an optimized vacuum sol deposition method. The phase composition, morphology, elemental characteristics, and chemical structure of uncoated and BGNs- and BGNs/GO-coated suture samples were characterized using Fourier transform infrared spectroscopy, field emission scanning electron microscopy, associated with elemental analysis, and knot performance test. In addition, in vitro bioactivity tests, biochemical tests, and in vivo tests were performed to examine the role of BGNs and GO on the biological and histopathological properties of the coated suture samples. The results indicated that the formation of BGNs and GO was enhanced significantly on the suture surface, which allowed for enhanced fibroblast attachment, migration, and proliferation and promoted the secretion of the angiogenic growth factor to speed up wound healing. These results confirmed the biocompatibility of BGNs- and BGNs/GO-coated suture samples and the positive effect of BGNs on the behavior of L929 fibroblast cells and also showed for the first time the possibility that cells can adhere and proliferate on the BGNs/GO-coated suture samples, especially in an in vivo environment. Resorbable surgical sutures with bioactive coatings, such as those prepared herein, can be an attractive biomaterial not only for hard tissue engineering but also for clinical applications in soft tissue engineering.
Collapse
Affiliation(s)
- Kerim Emre Öksüz
- Department
of Metallurgical and Materials Engineering, Faculty of Engineering, Sivas Cumhuriyet University, Sivas 58140, Türkiye
| | - Begüm Kurt
- Department
of Gynecology and Obstetrics, Faculty of Medicine Hospital, Sivas Cumhuriyet University, Sivas 58140, Türkiye
| | - Zeynep Deniz Şahin İnan
- Department
of Histology-Embryology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas 58140, Türkiye
| | - Ceylan Hepokur
- Department
of Biochemistry, Faculty of Pharmacy, Sivas
Cumhuriyet University, Sivas 58140, Türkiye
| |
Collapse
|
36
|
Zhang H, Lan D, Wu B, Chen X, Li X, Li Z, Dai F. Electrospun Piezoelectric Scaffold with External Mechanical Stimulation for Promoting Regeneration of Peripheral Nerve Injury. Biomacromolecules 2023. [PMID: 37329512 DOI: 10.1021/acs.biomac.3c00311] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Safe and efficient provision of electrical stimulation (ES) for nerve repair and regeneration is a problem that needs to be addressed. In this study, a silk fibroin/poly(vinylidene fluoride-co-hexafluoropropylene)/Ti3C2Tx (SF/PVDF-HFP/MXene) composite scaffold with piezoelectricity was developed by electrospinning technology. MXene was loaded to the scaffold to enhance the piezoelectric properties (Output voltage reaches up to 100 mV), mechanical properties, and antibacterial activity. Cell experiments demonstrated piezoelectric stimulation under external ultrasonication for promoting the growth and proliferation of Schwann cells (SCs) cultured on this electrospun scaffold. Further in vivo study with rat sciatic nerve injury model revealed that the SF/PVDF-HFP/MXene nerve conduit could induce the proliferation of SCs, enhance the elongation of axon, and promote axonal myelination. Under the piezoelectric effect of this nerve scaffold, the rats with regenerative nerve exhibited a favorable recovery effect of motor and sensory function, indicating a safe and feasible method of using this SF/PVDF-HFP/MXene piezoelectric scaffold for ES provision in vivo.
Collapse
Affiliation(s)
- Haiqiang Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Dongwei Lan
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Baiqing Wu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Xiang Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Xia Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Zhi Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China
| |
Collapse
|
37
|
Zhang Q, Chen J, Feng Y, Lin J, Li J, Wang Y, Tan H. Electroactive scaffolds of biodegradable polyurethane/polydopamine-functionalized graphene oxide regulating the inflammatory response and revitalizing the axonal growth cone for peripheral nerve regeneration. J Mater Chem B 2023. [PMID: 37326438 DOI: 10.1039/d3tb00837a] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Long-gap peripheral nerve injury remains a major challenge in regenerative medicine and results in permanent sensory and motor dysfunction. Nerve guidance scaffolds (NGSs) are known as a promising alternative to autologous nerve grafting. The latter, the current "gold standard" in clinical practice, is frequently constrained by the limited availability of sources and the inevitable damage to the donor area. Given the electrophysiological properties of nerves, electroactive biomaterials are being intensively investigated in nerve tissue engineering. In this study, we engineered a conductive NGS compounded of biodegradable waterborne polyurethane (WPU) and polydopamine-reduced graphene oxide (pGO) for repairing impaired peripheral nerves. The incorporation of pGO at the optimal concentration (3 wt%) promoted in vitro spreading of Schwann cells (SCs) with high expression of the proliferation marker S100 protein. In an in vivo study of sciatic nerve transection injury, WPU/pGO NGSs were found to regulate the immune microenvironment by activating macrophage M2 polarization and upregulate growth-associated protein 43 (GAP43) to facilitate axonal elongation. Histological and motor function analysis demonstrated that WPU/pGO NGSs had a neuroprosthetic effect close to that of an autograft, which significantly promoted the regeneration of myelinated axons, reduced gastrocnemius atrophy, and enhanced hindlimb motor function. These findings together suggested that electroactive WPU/pGO NGSs may represent a safe and effective strategy to manage large nerve defects.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, China.
| | - Jinlin Chen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuan Feng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jingjing Lin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yanchao Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610000, China.
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Med-X Center of Materials, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
38
|
Taşdemir Ş, Morçimen ZG, Doğan AA, Görgün C, Şendemir A. Surface Area of Graphene Governs Its Neurotoxicity. ACS Biomater Sci Eng 2023. [PMID: 37201186 DOI: 10.1021/acsbiomaterials.3c00104] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Due to their unique physicochemical properties, graphene and its derivatives are widely exploited for biomedical applications. It has been shown that graphene may exert different degrees of toxicity in in vivo or in vitro models when administered via different routes and penetrated through physiological barriers, subsequently being distributed within tissues or located within cells. In this study, in vitro neurotoxicity of graphene with different surface areas (150 and 750 m2/g) was examined on dopaminergic neuron model cells. SH-SY5Y cells were treated with graphene possessing two different surface areas (150 and 750 m2/g) in different concentrations between 400 and 3.125 μg/mL, and the cytotoxic and genotoxic effects were investigated. Both sizes of graphene have shown increased cell viability in decreasing concentrations. Cell damage increased with higher surface area. Lactate dehydrogenase (LDH) results have concluded that the viability loss of the cells is not through membrane damage. Neither of the two graphene types showed damage through lipid peroxidation (MDA) oxidative stress pathway. Glutathione (GSH) values increased within the first 24 and 48 h for both types of graphene. This increase suggests that graphene has an antioxidant effect on the SH-SY5Y model neurons. Comet analysis shows that graphene does not show genotoxicity on either surface area. Although there are many studies on graphene and its derivatives on their use with different cells in the literature, there are conflicting results in these studies, and most of the literature is focused on graphene oxide. Among these studies, no study examining the effect of graphene surface areas on the cell was found. Our study contributes to the literature in terms of examining the cytotoxic and genotoxic behavior of graphene with different surface areas.
Collapse
Affiliation(s)
- Şeyma Taşdemir
- Bioengineering Department, Celal Bayar University, Manisa 45140, Turkey
| | | | | | - Cansu Görgün
- Department of Experimental Medicine (DIMES), University of Genova, Genova 16126, Italy
| | - Aylin Şendemir
- Department of Bioengineering, Ege University, Izmir 35040, Turkey
- Department of Biomedical Technologies, Ege University, Izmir 35040, Turkey
| |
Collapse
|
39
|
Raghavan A, Radhakrishnan M, Soren K, Wadnerkar P, Kumar A, Chakravarty S, Ghosh S. Biological Evaluation of Graphene Quantum Dots and Nitrogen-Doped Graphene Quantum Dots as Neurotrophic Agents. ACS APPLIED BIO MATERIALS 2023. [PMID: 37167607 DOI: 10.1021/acsabm.3c00099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Over time, developments in nano-biomedical research have led to the creation of a number of systems to cure serious illnesses. Tandem use of nano-theragnostics such as diagnostic and therapeutic approaches tailored to the individual disease treatment is crucial for further development in the field of biomedical advancements. Graphene has garnered attention in the recent times as a potential nanomaterial for tissue engineering and regenerative medicines owing to its biocompatibility among the several other unique properties it possesses. The zero-dimensional graphene quantum dots (GQDs) and their nitrogen-doped variant, nitrogen-doped GQDs (N-GQDs), have good biocompatibility, and optical and physicochemical properties. GQDs have been extensively researched owing to several factors such as their size, surface charge, and interactions with other molecules found in biological media. This work briefly elucidates the potential of electroactive GQDs as well as N-GQDs as neurotrophic agents. In vitro investigations employing the N2A cell line were used to evaluate the effectiveness of GQDs and N-GQDs as neurotrophic agents, wherein basic investigations such as SRB assay and neurite outgrowth assay were performed. The results inferred from immunohistochemistry followed by confocal imaging studies as well as quantitative real-time PCR (qPCR) studies corroborated those obtained from neurite outgrowth assay. We have also conducted a preliminary investigation of the pattern of gene expression for neurotrophic and gliotrophic growth factors using ex vivo neuronal and mixed glial cultures taken from the brains of postnatal day 2 mice pups. Overall, the studies indicated that GQDs and N-GQDs hold prospect as a framework for further development of neuroactive compounds for relevant central nervous system (CNS) purposes.
Collapse
Affiliation(s)
- Akshaya Raghavan
- Polymers & Functional Materials Division, CSIR─Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mydhili Radhakrishnan
- Applied Biology Division, CSIR─Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kalyani Soren
- Applied Biology Division, CSIR─Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | - Arvind Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- CSIR─Centre for Cellular and Molecular Biology, Hyderabad 500007, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR─Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sutapa Ghosh
- Polymers & Functional Materials Division, CSIR─Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
40
|
Fang Y, Wang C, Liu Z, Ko J, Chen L, Zhang T, Xiong Z, Zhang L, Sun W. 3D Printed Conductive Multiscale Nerve Guidance Conduit with Hierarchical Fibers for Peripheral Nerve Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205744. [PMID: 36808712 PMCID: PMC10131803 DOI: 10.1002/advs.202205744] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Nerve guidance conduits (NGCs) have become a promising alternative for peripheral nerve regeneration; however, the outcome of nerve regeneration and functional recovery is greatly affected by the physical, chemical, and electrical properties of NGCs. In this study, a conductive multiscale filled NGC (MF-NGC) consisting of electrospun poly(lactide-co-caprolactone) (PCL)/collagen nanofibers as the sheath, reduced graphene oxide /PCL microfibers as the backbone, and PCL microfibers as the internal structure for peripheral nerve regeneration is developed. The printed MF-NGCs presented good permeability, mechanical stability, and electrical conductivity, which further promoted the elongation and growth of Schwann cells and neurite outgrowth of PC12 neuronal cells. Animal studies using a rat sciatic nerve injury model reveal that the MF-NGCs promote neovascularization and M2 transition through the rapid recruitment of vascular cells and macrophages. Histological and functional assessments of the regenerated nerves confirm that the conductive MF-NGCs significantly enhance peripheral nerve regeneration, as indicated by improved axon myelination, muscle weight increase, and sciatic nerve function index. This study demonstrates the feasibility of using 3D-printed conductive MF-NGCs with hierarchically oriented fibers as functional conduits that can significantly enhance peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Chengjin Wang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Zibo Liu
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Jeonghoon Ko
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Li Chen
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Ting Zhang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Zhuo Xiong
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Lei Zhang
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
| | - Wei Sun
- Biomanufacturing CenterDepartment of Mechanical EngineeringTsinghua UniversityBeijing100084P. R. China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of BeijingBeijing100084P. R. China
- “Biomanufacturing and Engineering Living Systems” Innovation International Talents Base (111 Base)Beijing100084P. R. China
- Department of Mechanical EngineeringDrexel UniversityPhiladelphiaPA19104USA
| |
Collapse
|
41
|
Wang W, Liu M, Shafiq M, Li H, Hashim R, El-Newehy M, El-Hamshary H, Morsi Y, Mo X. Synthesis of oxidized sodium alginate and its electrospun bio-hybrids with zinc oxide nanoparticles to promote wound healing. Int J Biol Macromol 2023; 232:123480. [PMID: 36720331 DOI: 10.1016/j.ijbiomac.2023.123480] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/22/2023] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Abstract
Electrospun fibers provide a promising platform for wound healing; however, they lack requisite characteristics for wound repair, including antibacterial and anti-inflammatory properties and angiogenic ability. Sodium alginate (SA) is being used for different types of applications. However, the poor spinnability of SA restricts its applications. The objectives of this study were three-fold: a) to synthesize oxidized sodium alginate (OSA) to improve its spinnability, b) to fabricate composite fibrous membranes by blending OSA along with zinc oxide nanoparticles (ZnO-NPs), and c) to decipher antibacterial and anti-inflammatory properties as well as biocompatibility of membranes in vitro and in vivo. OSA displaying different oxidation degrees (Dox (%)) was synthesized by varying the molar ratio of sodium periodate to SA. OSA (Dox, ∼48 %) afforded smooth and uniform fibers; 0.5 wt% of adipic dihydrazide (ADH) evolved into structurally stable and water-insoluble membranes. Composite fibrous membranes containing 2 wt% of ZnO-NPs displayed good biocompatibility and bactericidal effect against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) in vitro. In addition, composite membranes showed remarkable epithelialization, neovascularization, and anti-inflammatory response than that of the membranes devoid of ZnO-NPs. Conclusively, these composite fibrous membranes may have broad implications for wound healing applications.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - MingYue Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Muhammad Shafiq
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China; Department of Chemical Engineering, Faculty of Chemical Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, Japan.
| | - HaiYan Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Rashida Hashim
- School of Physical Sciences, University of Punjab (PU), Lahore 54000, Pakistan
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Hany El-Hamshary
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Yosry Morsi
- Faculty of Engineering and Industrial Sciences, Swinburne University of Technology, Boroondara, VIC 3122, Australia
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
42
|
Barroca N, da Silva DM, Pinto SC, Sousa JPM, Verstappen K, Klymov A, Fernández-San-Argimiro FJ, Madarieta I, Murua O, Olalde B, Papadimitriou L, Karali K, Mylonaki K, Stratakis E, Ranella A, Marques PAAP. Interfacing reduced graphene oxide with an adipose-derived extracellular matrix as a regulating milieu for neural tissue engineering. BIOMATERIALS ADVANCES 2023; 148:213351. [PMID: 36842343 DOI: 10.1016/j.bioadv.2023.213351] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Enthralling evidence of the potential of graphene-based materials for neural tissue engineering is motivating the development of scaffolds using various structures related to graphene such as graphene oxide (GO) or its reduced form. Here, we investigated a strategy based on reduced graphene oxide (rGO) combined with a decellularized extracellular matrix from adipose tissue (adECM), which is still unexplored for neural repair and regeneration. Scaffolds containing up to 50 wt% rGO relative to adECM were prepared by thermally induced phase separation assisted by carbodiimide (EDC) crosslinking. Using partially reduced GO enables fine-tuning of the structural interaction between rGO and adECM. As the concentration of rGO increased, non-covalent bonding gradually prevailed over EDC-induced covalent conjugation with the adECM. Edge-to-edge aggregation of rGO favours adECM to act as a biomolecular physical crosslinker to rGO, leading to the softening of the scaffolds. The unique biochemistry of adECM allows neural stem cells to adhere and grow. Importantly, high rGO concentrations directly control cell fate by inducing the differentiation of both NE-4C cells and embryonic neural progenitor cells into neurons. Furthermore, primary astrocyte fate is also modulated as increasing rGO boosts the expression of reactivity markers while unaltering the expression of scar-forming ones.
Collapse
Affiliation(s)
- Nathalie Barroca
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal.
| | - Daniela M da Silva
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal
| | - Susana C Pinto
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal
| | - Joana P M Sousa
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal
| | - Kest Verstappen
- Radboud University Nijmegen Medical Centre, Department of Regenerative Biomaterials, 6500HB Nijmegen, the Netherlands
| | - Alexey Klymov
- Radboud University Nijmegen Medical Centre, Department of Regenerative Biomaterials, 6500HB Nijmegen, the Netherlands
| | | | - Iratxe Madarieta
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Olatz Murua
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Beatriz Olalde
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Lina Papadimitriou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Kanelina Karali
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Konstantina Mylonaki
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Anthi Ranella
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece.
| | - Paula A A P Marques
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal.
| |
Collapse
|
43
|
Tan Y, Chen Y, Lu T, Witman N, Yan B, Gong Y, Ai X, Yang L, Liu M, Luo R, Wang H, Ministrini S, Dong W, Wang W, Fu W. Engineering a conduction-consistent cardiac patch with rGO/PLCL electrospun nanofibrous membranes and human iPSC-derived cardiomyocytes. Front Bioeng Biotechnol 2023; 11:1094397. [PMID: 36845196 PMCID: PMC9944832 DOI: 10.3389/fbioe.2023.1094397] [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: 11/10/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
The healthy human heart has special directional arrangement of cardiomyocytes and a unique electrical conduction system, which is critical for the maintenance of effective contractions. The precise arrangement of cardiomyocytes (CMs) along with conduction consistency between CMs is essential for enhancing the physiological accuracy of in vitro cardiac model systems. Here, we prepared aligned electrospun rGO/PLCL membranes using electrospinning technology to mimic the natural heart structure. The physical, chemical and biocompatible properties of the membranes were rigorously tested. We next assembled human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on electrospun rGO/PLCL membranes in order to construct a myocardial muscle patch. The conduction consistency of cardiomyocytes on the patches were carefully recorded. We found that cells cultivated on the electrospun rGO/PLCL fibers presented with an ordered and arranged structure, excellent mechanical properties, oxidation resistance and effective guidance. The addition of rGO was found to be beneficial for the maturation and synchronous electrical conductivity of hiPSC-CMs within the cardiac patch. This study verified the possibility of using conduction-consistent cardiac patches to enhance drug screening and disease modeling applications. Implementation of such a system could one day lead to in vivo cardiac repair applications.
Collapse
Affiliation(s)
- Yao Tan
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Chen
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Lu
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nevin Witman
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Bingqian Yan
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yiqi Gong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefeng Ai
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Yang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Minglu Liu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Runjiao Luo
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huijing Wang
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland,Department of Medicine and Surgery, Internal Medicine, Angiology and Atherosclerosis, University of Perugia, Perugia, Italy
| | - Wei Dong
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| | - Wei Wang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,Shanghai Key Laboratory of Tissue Engineering, Shanghai 9th People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Wei Dong, ; Wei Wang, ; Wei Fu,
| |
Collapse
|
44
|
Lu S, Chen W, Wang J, Guo Z, Xiao L, Wei L, Yu J, Yuan Y, Chen W, Bian M, Huang L, Liu Y, Zhang J, Li YL, Jiang LB. Polydopamine-Decorated PLCL Conduit to Induce Synergetic Effect of Electrical Stimulation and Topological Morphology for Peripheral Nerve Regeneration. SMALL METHODS 2023; 7:e2200883. [PMID: 36596669 DOI: 10.1002/smtd.202200883] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/09/2022] [Indexed: 06/17/2023]
Abstract
Due to the limited self-repairing capacity after peripheral nerve injuries (PNI), artificial nerve conduits are widely applied to facilitate neural regeneration. Exogenous electrical stimulation (ES) that is carried out by the conductive conduit regulates the biological behavior of Schwann cells (SCs). Meanwhile, a longitudinal surface structure counts to guide axonal growth to accelerate the end-to-end connection. Currently, there are no conduits equipped with both electrical conduction and axon-guiding surface structure. Herein, a biodegradable, conductive poly(l-lactide-co-caprolactone)/graphene (PLCL/GN) composite conduit is designed. The conduit with 20.96 ± 1.26 MPa tensile strength has a micropatterned surface of 20 µm groove fabricated by microimprint technology and self-assembled polydopamine (PDA). In vitro evaluation shows that the conduits with ES effectively stimulate the directional cell migration, adhesion, and elongation, and enhance neuronal expression of SCs. The rat sciatic nerve crush model demonstrates that the conductive micropatterned conduit with ES promotes the growth of myelin sheath, faster nerve regeneration, and 20-fold functional recovery in vivo. These discoveries prove that the PLCL(G)/PDA/GN composite conduit is a promising tool for PNI treatment by providing the functional integration of physical guidance, biomimetic biological regulation, and bioelectrical stimulation, which inspires a novel therapeutic approach for nerve regeneration in the future.
Collapse
Affiliation(s)
- Shunyi Lu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Chen
- The Key Laboratory for Ultrafine Materials of Ministry of Education, State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jiayi Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zilong Guo
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, 4059, Australia
| | - Lingyu Wei
- The Key Laboratory for Ultrafine Materials of Ministry of Education, State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jieqin Yu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ya Yuan
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weisin Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Mengxuan Bian
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Huang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Jian Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu-Lin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| | - Li-Bo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
45
|
Jalilinejad N, Rabiee M, Baheiraei N, Ghahremanzadeh R, Salarian R, Rabiee N, Akhavan O, Zarrintaj P, Hejna A, Saeb MR, Zarrabi A, Sharifi E, Yousefiasl S, Zare EN. Electrically conductive carbon-based (bio)-nanomaterials for cardiac tissue engineering. Bioeng Transl Med 2023; 8:e10347. [PMID: 36684103 PMCID: PMC9842069 DOI: 10.1002/btm2.10347] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
A proper self-regenerating capability is lacking in human cardiac tissue which along with the alarming rate of deaths associated with cardiovascular disorders makes tissue engineering critical. Novel approaches are now being investigated in order to speedily overcome the challenges in this path. Tissue engineering has been revolutionized by the advent of nanomaterials, and later by the application of carbon-based nanomaterials because of their exceptional variable functionality, conductivity, and mechanical properties. Electrically conductive biomaterials used as cell bearers provide the tissue with an appropriate microenvironment for the specific seeded cells as substrates for the sake of protecting cells in biological media against attacking mechanisms. Nevertheless, their advantages and shortcoming in view of cellular behavior, toxicity, and targeted delivery depend on the tissue in which they are implanted or being used as a scaffold. This review seeks to address, summarize, classify, conceptualize, and discuss the use of carbon-based nanoparticles in cardiac tissue engineering emphasizing their conductivity. We considered electrical conductivity as a key affecting the regeneration of cells. Correspondingly, we reviewed conductive polymers used in tissue engineering and specifically in cardiac repair as key biomaterials with high efficiency. We comprehensively classified and discussed the advantages of using conductive biomaterials in cardiac tissue engineering. An overall review of the open literature on electroactive substrates including carbon-based biomaterials over the last decade was provided, tabulated, and thoroughly discussed. The most commonly used conductive substrates comprising graphene, graphene oxide, carbon nanotubes, and carbon nanofibers in cardiac repair were studied.
Collapse
Affiliation(s)
- Negin Jalilinejad
- Biomaterial Group, Department of Biomedical EngineeringAmirkabir University of TechnologyTehranIran
| | - Mohammad Rabiee
- Biomaterial Group, Department of Biomedical EngineeringAmirkabir University of TechnologyTehranIran
| | - Nafiseh Baheiraei
- Tissue Engineering and Applied Cell Sciences Division, Department of Anatomical Sciences, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | | | - Reza Salarian
- Biomedical Engineering DepartmentMaziar UniversityRoyanMazandaranIran
| | - Navid Rabiee
- Department of PhysicsSharif University of TechnologyTehranIran
- School of EngineeringMacquarie UniversitySydneyNew South WalesAustralia
- Department of Materials Science and EngineeringPohang University of Science and Technology (POSTECH), 77 Cheongam‐ro, Nam‐guPohangGyeongbukSouth Korea
| | - Omid Akhavan
- Department of PhysicsSharif University of TechnologyTehranIran
| | - Payam Zarrintaj
- School of Chemical EngineeringOklahoma State UniversityStillwaterOklahomaUSA
| | - Aleksander Hejna
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural SciencesIstinye UniversityIstanbulTurkey
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and TechnologiesHamadan University of Medical SciencesHamadanIran
| | - Satar Yousefiasl
- School of DentistryHamadan University of Medical SciencesHamadanIran
| | | |
Collapse
|
46
|
Han N, Zhang W, Fang XX, Li QC, Pi W. Reduced graphene oxide-embedded nerve conduits loaded with bone marrow mesenchymal stem cell-derived extracellular vesicles promote peripheral nerve regeneration. Neural Regen Res 2023. [PMID: 35799543 PMCID: PMC9241414 DOI: 10.4103/1673-5374.343889] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We previously combined reduced graphene oxide (rGO) with gelatin-methacryloyl (GelMA) and polycaprolactone (PCL) to create an rGO-GelMA-PCL nerve conduit and found that the conductivity and biocompatibility were improved. However, the rGO-GelMA-PCL nerve conduits differed greatly from autologous nerve transplants in their ability to promote the regeneration of injured peripheral nerves and axonal sprouting. Extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSCs) can be loaded into rGO-GelMA-PCL nerve conduits for repair of rat sciatic nerve injury because they can promote angiogenesis at the injured site. In this study, 12 weeks after surgery, sciatic nerve function was measured by electrophysiology and sciatic nerve function index, and myelin sheath and axon regeneration were observed by electron microscopy, immunohistochemistry, and immunofluorescence. The regeneration of microvessel was observed by immunofluorescence. Our results showed that rGO-GelMA-PCL nerve conduits loaded with BMSC-derived extracellular vesicles were superior to rGO-GelMA-PCL conduits alone in their ability to increase the number of newly formed vessels and axonal sprouts at the injury site as well as the recovery of neurological function. These findings indicate that rGO-GelMA-PCL nerve conduits loaded with BMSC-derived extracellular vesicles can promote peripheral nerve regeneration and neurological function recovery, and provide a new direction for the curation of peripheral nerve defect in the clinic.
Collapse
|
47
|
Zhang H, Lan D, Li X, Li Z, Dai F. Conductive and antibacterial scaffold with rapid crimping property for application prospect in repair of peripheral nerve injury. J Appl Polym Sci 2022. [DOI: 10.1002/app.53426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Hai‐qiang Zhang
- State Key Laboratory of Silkworm Genome Biology Southwest University Chongqing China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences Southwest University Chongqing China
| | - Dong‐wei Lan
- State Key Laboratory of Silkworm Genome Biology Southwest University Chongqing China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences Southwest University Chongqing China
| | - Xia Li
- State Key Laboratory of Silkworm Genome Biology Southwest University Chongqing China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences Southwest University Chongqing China
| | - Zhi Li
- State Key Laboratory of Silkworm Genome Biology Southwest University Chongqing China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences Southwest University Chongqing China
| | - Fang‐Yin Dai
- State Key Laboratory of Silkworm Genome Biology Southwest University Chongqing China
- Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of sericulture, Textile and Biomass Sciences Southwest University Chongqing China
- Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs Southwest University Chongqing China
| |
Collapse
|
48
|
Vuković JS, Filipović VV, Babić Radić MM, Vukomanović M, Milivojevic D, Ilic-Tomic T, Nikodinovic-Runic J, Tomić SL. In Vitro and In Vivo Biocompatible and Controlled Resveratrol Release Performances of HEMA/Alginate and HEMA/Gelatin IPN Hydrogel Scaffolds. Polymers (Basel) 2022; 14:polym14204459. [PMID: 36298041 PMCID: PMC9610835 DOI: 10.3390/polym14204459] [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: 08/28/2022] [Revised: 10/02/2022] [Accepted: 10/17/2022] [Indexed: 01/19/2023] Open
Abstract
Scaffold hydrogel biomaterials designed to have advantageous biofunctional properties, which can be applied for controlled bioactive agent release, represent an important concept in biomedical tissue engineering. Our goal was to create scaffolding materials that mimic living tissue for biomedical utilization. In this study, two novel series of interpenetrating hydrogel networks (IPNs) based on 2-hydroxyethyl methacrylate/gelatin and 2-hydroxyethyl methacrylate/alginate were crosslinked using N-ethyl-N'-(3-dimethyl aminopropyl)carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS). Characterization included examining the effects of crosslinker type and concentration on structure, morphological and mechanical properties, in vitro swelling, hydrophilicity as well as on the in vitro cell viability (fibroblast cells) and in vivo (Caenorhabditis elegans) interactions of novel biomaterials. The engineered IPN hydrogel scaffolds show an interconnected pore morphology and porosity range of 62.36 to 85.20%, favorable in vitro swelling capacity, full hydrophilicity, and Young's modulus values in the range of 1.40 to 7.50 MPa. In vitro assay on healthy human fibroblast (MRC5 cells) by MTT test and in vivo (Caenorhabditis elegans) survival assays show the advantageous biocompatible properties of novel IPN hydrogel scaffolds. Furthermore, in vitro controlled release study of the therapeutic agent resveratrol showed that these novel scaffolding systems are suitable controlled release platforms. The results revealed that the use of EDC and the combination of EDC/NHS crosslinkers can be applied to prepare and tune the properties of the IPN 2-hydroxyethyl methacrylate/alginate and 2-hydroxyethyl methacrylate/gelatin hydrogel scaffolds series, which have shown great potential for biomedical engineering applications.
Collapse
Affiliation(s)
- Jovana S. Vuković
- University of Belgrade, Faculty of Technology and Metallurgy, Karnegijeva 4, 11000 Belgrade, Serbia
| | - Vuk V. Filipović
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Vojvode Stepe 444a, 11000 Belgrade, Serbia
| | - Marija M. Babić Radić
- University of Belgrade, Faculty of Technology and Metallurgy, Karnegijeva 4, 11000 Belgrade, Serbia
| | - Marija Vukomanović
- Advanced Materials Department, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia
| | - Dusan Milivojevic
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Vojvode Stepe 444a, 11000 Belgrade, Serbia
| | - Tatjana Ilic-Tomic
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Vojvode Stepe 444a, 11000 Belgrade, Serbia
| | - Jasmina Nikodinovic-Runic
- University of Belgrade, Institute of Molecular Genetics and Genetic Engineering, Vojvode Stepe 444a, 11000 Belgrade, Serbia
| | - Simonida Lj. Tomić
- University of Belgrade, Faculty of Technology and Metallurgy, Karnegijeva 4, 11000 Belgrade, Serbia
- Correspondence: ; Tel.: +381-11-3303-630
| |
Collapse
|
49
|
Zou S, Yao X, Shao H, Reis RL, Kundu SC, Zhang Y. Nonmulberry silk fibroin-based biomaterials: Impact on cell behavior regulation and tissue regeneration. Acta Biomater 2022; 153:68-84. [PMID: 36113722 DOI: 10.1016/j.actbio.2022.09.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/28/2022] [Accepted: 09/08/2022] [Indexed: 11/01/2022]
Abstract
Silk fibroin (SF) is a promising biomaterial due to its good biocompatibility, easy availability, and high mechanical properties. Compared with mulberry silk fibroin (MSF), nonmulberry silk fibroin (NSF) isolated from typical nonmulberry silkworm silk exhibits unique arginine-glycine-aspartic acid (RGD) sequences with favorable cell adhesion enhancing effect. This inherent property probably makes the NSF more suitable for cell culture and tissue regeneration-related applications. Accordingly, various types of NSF-based biomaterials, such as particles, films, fiber mats, and 3D scaffolds, are constructed and their application potential in different biomedical fields is extensively investigated. Based on these promising NSF biomaterials, this review firstly makes a systematical comparison between the molecular structure and properties of MSF and typical NSF and highlights the unique properties of NSF. In addition, we summarize the effective fabrication strategies from degummed nonmulberry silk fibers to regenerated NSF-based biomaterials with controllable formats and their recent application progresses in cell behavior regulation and tissue regeneration. Finally, current challenges and future perspectives for the fabrication and application of NSF-based biomaterials are discussed. Related research and perspectives may provide valuable references for designing and modifying effective NSF-based and other natural biomaterials. STATEMENT OF SIGNIFICANCE: There exist many reviews about mulberry silk fibroin (MSF) biomaterials and their biomedical applications, while that about nonmulberry silk fibroin (NSF) biomaterials is scarce. Compared with MSF, NSF exhibits unique arginine-glycine-aspartic acid sequences with promising cell adhesion enhancing effect, which makes NSF more suitable for cell culture and tissue regeneration related applications. Focusing on these advanced NSF biomaterials, this review has systematically compared the structure and properties of MSF and NSF, and emphasized the unique properties of NSF. Following that, the effective construction strategies for NSF-based biomaterials are summarized, and their recent applications in cell behavior regulations and tissue regenerations are highlighted. Furthermore, current challenges and future perspectives for the fabrication and application of NSF-based biomaterials were discussed.
Collapse
Affiliation(s)
- Shengzhi Zou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Xiang Yao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Huili Shao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Rui L Reis
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco, Guimarães 4805-017, Portugal
| | - Subhas C Kundu
- I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco, Guimarães 4805-017, Portugal
| | - Yaopeng Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Materials Science and Engineering, Donghua University, Shanghai 201620, People's Republic of China.
| |
Collapse
|
50
|
Maeng WY, Tseng WL, Li S, Koo J, Hsueh YY. Electroceuticals for peripheral nerve regeneration. Biofabrication 2022; 14. [PMID: 35995036 PMCID: PMC10109522 DOI: 10.1088/1758-5090/ac8baa] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/22/2022] [Indexed: 11/12/2022]
Abstract
Electroceuticals provide promising opportunities for peripheral nerve regeneration, in terms of modulating the extensive endogenous tissue repair mechanisms between neural cell body, axons and target muscles. However, great challenges remain to deliver effective and controllable electroceuticals via bioelectronic implantable device. In this review, the modern fabrication methods of bioelectronic conduit for bridging critical nerve gaps after nerve injury are summarized, with regard to conductive materials and core manufacturing process. In addition, to deliver versatile electrical stimulation, the integration of implantable bioelectronic device is discussed, including wireless energy harvesters, actuators and sensors. Moreover, a comprehensive insight of beneficial mechanisms is presented, including up-to-date in vitro, in vivo and clinical evidence. By integrating conductive biomaterials, 3D engineering manufacturing process and bioelectronic platform to deliver versatile electroceuticals, the modern biofabrication enables comprehensive biomimetic therapies for neural tissue engineering and regeneration in the new era.
Collapse
Affiliation(s)
- Woo-Youl Maeng
- Bio-Medical Engineering, Korea University, B156, B, Hana Science Hall, 145, Anam-ro, Seongbuk-gu, Seoul, Seongbuk-gu, Seoul, 02841, Korea (the Republic of)
| | - Wan Ling Tseng
- Department of Surgery, National Cheng Kung University College of Medicine, No.138, Sheng-Li road, Tainan, 701, TAIWAN
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, 5121 Eng V, Los Angeles, California, 90095, UNITED STATES
| | - Jahyun Koo
- Biomedical Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, 02841, Korea (the Republic of)
| | - Yuan-Yu Hsueh
- Department of Surgery, National Cheng Kung University College of Medicine, No.138, Sheng-Li road, Tainan, 701, TAIWAN
| |
Collapse
|