1
|
Ma Q, Meng K, Zhang KQ, Ramakrishna S, Zhao H. A critical view of silk fibroin for non-viral gene therapy. Int J Biol Macromol 2025; 298:139817. [PMID: 39818397 DOI: 10.1016/j.ijbiomac.2025.139817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Exogenous genes are inserted into target cells during gene therapy in order to compensate or rectify disorders brought on by faulty or aberrant genes. However, gene therapy is still in its early stages because of its unsatisfactory therapeutic effects which are mainly due to low transfection efficiency of vectors, high toxicity, and poor target specificity. A natural polymer with numerous bioactive sites, good mechanical qualities, biodegradability, biocompatibility, and processability called silk fibroin has gained attention as a possible gene therapy vector. Using silk fibroin as a gene vector can reduce cell toxicity, extend the duration of gene expression, and allow further release even in the bloodstream, thereby expanding its therapeutic scope. This review outlines the advancements made with regard to gene delivery methods based on silk fibroin materials in the fields of malignant tumors, bone tissue regeneration, neural tissue, and vascular tissue engineering. Silk fibroin exhibits remarkable repair and therapeutic effects in gene therapy and can be employed in numerous forms, such as a vector (nanoparticles, microcapsules) or a matrix (hydrogel, scaffold) for gene delivery.
Collapse
Affiliation(s)
- Qirui Ma
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, PR China
| | - Kai Meng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, PR China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, PR China
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, College of Design and Engineering, National University of Singapore, 9 Engineering Drive 1, #07-08 Block EA, Singapore 117575, Singapore
| | - Huijing Zhao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, No. 199 Ren'ai Road, Industrial Park, Suzhou 215123, PR China.
| |
Collapse
|
2
|
Widodo W, Aprilya D, Satria O. Regenerative Medicine: A New Horizon in Peripheral Nerve Injury and Repair. Orthop Rev (Pavia) 2025; 17:133572. [PMID: 40176924 PMCID: PMC11964392 DOI: 10.52965/001c.133572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 12/21/2024] [Indexed: 04/05/2025] Open
Abstract
A peripheral nerve injury is a great burden for the patient and a challenge for the clinician. In a complete injury (axonotmesis or neurotmesis), the slow nature of nerve regeneration after repair or reconstruction hardly catches up to the target organ's degeneration rate, leading to a poor prognosis. The current advance in regenerative medicine has shown the potency of stem cells and their products for healing many human body structures, including the nerve. A comprehensive literature search was conducted using an internet-based search engine for current advances in regenerative medicine to augment peripheral nerve repair or reconstruction. Stem cells can differentiate into nerve cells and have paracrine and immunomodulatory effects. Its products, such as the secretome and exosome, have also been studied, and they have many benefits for the regeneration process. This novel treatment possesses significant potential to accelerate nerve healing after nerve reconstruction and potentially postpone the degenerative process in the target organ, allowing it to respond to the new signal once nerve regeneration is complete. The aim of this article is to summarized the application of stem cells and its products for nerve healing.
Collapse
Affiliation(s)
- Wahyu Widodo
- Hand and Microsurgery Division, Orthopedic and Traumatology Department, Fatmawati General Hospital, Jakarta, Indonesia
| | - Dina Aprilya
- Hand and Microsurgery Division, Orthopedic and Traumatology Department, Fatmawati General Hospital, Jakarta, Indonesia
| | - Oryza Satria
- Hand and Microsurgery Division, Orthopedic and Traumatology Department, Fatmawati General Hospital, Jakarta, Indonesia
| |
Collapse
|
3
|
Tsai Y, Song J, Shi R, Knöll B, Synatschke CV. A Roadmap of Peptide-Based Materials in Neural Regeneration. Adv Healthc Mater 2025; 14:e2402939. [PMID: 39540310 PMCID: PMC11730414 DOI: 10.1002/adhm.202402939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Injuries to the nervous system lead to irreversible damage and limited functional recovery. The peripheral nervous system (PNS) can self-regenerate to some extent for short nerve gaps. In contrast, the central nervous system (CNS) has an intrinsic limitation to self-repair owing to its convoluted neural microenvironment and inhibitory response. The primary phase of CNS injury, happening within 48 h, results from external impacts like mechanical stress. Afterward, the secondary phase of the injury occurs, originating from neuronal excitotoxicity, mitochondrial dysfunction, and neuroinflammation. No golden standard to treat injured neurons exists, and conventional medicine serves only as a protective approach to alleviating the symptoms of chronic injury. Synthetic peptides provide a promising approach for neural repair, either as soluble drugs or by using their intrinsic self-assembly propensity to serve as an extracellular matrix (ECM) mimic for cell adhesion and to incorporate bioactive epitopes. In this review, an overview of nerve injury models, common in vitro models, and peptide-based therapeutics such as ECM mimics is provided. Due to the complexity of treating neuronal injuries, a multidisciplinary collaboration between biologists, physicians, and material scientists is paramount. Together, scientists with complementary expertise will be required to formulate future therapeutic approaches for clinical use.
Collapse
Affiliation(s)
- Yu‐Liang Tsai
- Department for Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jialei Song
- Institute of NeurobiochemistryUniversity of UlmAlbert‐Einstein‐Allee 11D‐89081UlmGermany
- Department of OrthopedicsShanghai 9th People's HospitalShanghai Jiao Tong University School of MedicineZhizaoju Road 639Shanghai200011China
| | - Rachel Shi
- Department for Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Yale School of Medicine333 Cedar StNew HavenCT06510USA
| | - Bernd Knöll
- Institute of NeurobiochemistryUniversity of UlmAlbert‐Einstein‐Allee 11D‐89081UlmGermany
| | - Christopher V. Synatschke
- Department for Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| |
Collapse
|
4
|
Salehpour A, Karimi Z, Ghasemi Zadeh M, Afshar M, Kameli A, Mooseli F, Zare M, Afshar A. Therapeutic potential of mesenchymal stem cell-derived exosomes and miRNAs in neuronal regeneration and rejuvenation in neurological disorders: a mini review. Front Cell Neurosci 2024; 18:1427525. [PMID: 39429946 PMCID: PMC11486650 DOI: 10.3389/fncel.2024.1427525] [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: 05/03/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have gained considerable attention in the field of regenerative medicine due to their ability to secrete small extracellular vesicles (EVs) known as exosomes. This review delves into the various biological activities of MSCs and the cell interactions enabled by these exosomes, with a focus on their potential for neuronal regeneration and the treatment of neurological disorders. We scrutinize findings from multiple studies that underscore the neuroprotective and neuro-regenerative effects of exosomes derived from MSCs, illuminating their mechanisms of action and therapeutic applications. This review thoroughly investigates all related pathways, miRNAs, and factors to suggest potential strategies for enhancing therapy for neurological disorders using exosomes and miRNAs, and for boosting neuronal regeneration.
Collapse
Affiliation(s)
- Aria Salehpour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zahra Karimi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mokhtar Ghasemi Zadeh
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Afshar
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ali Kameli
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Fatemeh Mooseli
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Masoud Zare
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Alireza Afshar
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
5
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
6
|
Aisaiti A, Aierxiding S, Shoukeer K, Muheremu A. Mesenchymal stem cells for peripheral nerve injury and regeneration: a bibliometric and visualization study. Front Neurol 2024; 15:1420402. [PMID: 39161869 PMCID: PMC11330774 DOI: 10.3389/fneur.2024.1420402] [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: 04/20/2024] [Accepted: 06/10/2024] [Indexed: 08/21/2024] Open
Abstract
Objective To use bibliometric methods to analyze the research hotspots and future development trends regarding the application of mesenchymal stem cells in peripheral nerve injury and regeneration. Methods Articles published from January 1, 2013, to December 31, 2023, were meticulously screened using the MeSH terms: TS = ("Mesenchymal stem cells" AND "Peripheral nerve injury") OR TS = ("Mesenchymal stem cells" AND "Peripheral nerve regeneration") within the Web of Science database. The compiled data was then subjected to in-depth analysis with the aid of VOSviewer and Cite Space software, which facilitated the identification of the most productive countries, organizations, authors, and the predominant keywords prevalent within this research domain. Results An extensive search of the Web of Science database yielded 350 relevant publications. These scholarly works were authored by 2,049 collaborative researchers representing 41 countries and affiliated with 585 diverse academic and research institutions. The findings from this research were disseminated across 167 various journals, and the publications collectively cited 21,064 references from 3,339 distinct journals. Conclusion Over the past decade, there has been a consistent upward trajectory in the number of publications and citations pertaining to the use of mesenchymal stem cells in the realm of peripheral nerve injury and regeneration. The domain of stem cell therapy for nerve injury has emerged as a prime focus of research, with mesenchymal stem cell therapy taking center stage due to its considerable promise in the treatment of nerve injuries. This therapeutic approach holds the potential to significantly enhance treatment options and rehabilitation prospects for patients suffering from such injuries.
Collapse
Affiliation(s)
- Aikebaierjiang Aisaiti
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Shalayiding Aierxiding
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Kutiluke Shoukeer
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Aikeremujiang Muheremu
- Key Laboratory of Orthopedic Regenerative Medicine, Sixth Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
- Beijing Darwin Cell Biotechnology Co., Ltd., Beijing, China
| |
Collapse
|
7
|
Li X, Xu H, Li C, Guan Y, Liu Y, Zhang T, Meng F, Cheng H, Song X, Jia Z, He R, Zhao J, Chen S, Guan C, Yan S, Wang J, Wei Y, Zhang J, Tang J, Peng J, Wang Y. Biological characteristics of tissue engineered-nerve grafts enhancing peripheral nerve regeneration. Stem Cell Res Ther 2024; 15:215. [PMID: 39020413 PMCID: PMC11256578 DOI: 10.1186/s13287-024-03827-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND A favorable regenerative microenvironment is essential for peripheral nerve regeneration. Neural tissue-specific extracellular matrix (ECM) is a natural material that helps direct cell behavior and promote axon regeneration. Both bone marrow-derived mesenchymal stem cells (BMSCs) and adipose-derived mesenchymal stem cells (ADSCs) transplantation are effective in repairing peripheral nerve injury (PNI). However, there is no study that characterizes the in vivo microenvironmental characteristics of these two MSCs for the early repair of PNI when combined with neural tissue-derived ECM materials, i.e., acellular nerve allograft (ANA). METHODS In order to investigate biological characteristics, molecular mechanisms of early stage, and effectiveness of ADSCs- or BMSCs-injected into ANA for repairing PNI in vivo, a rat 10 mm long sciatic nerve defect model was used. We isolated primary BMSCs and ADSCs from bone marrow and adipose tissue, respectively. First, to investigate the in vivo response characteristics and underlying molecular mechanisms of ANA combined with BMSCs or ADSCs, eighty-four rats were randomly divided into three groups: ANA group, ANA+BMSC group, and ANA+ADSC group. We performed flow cytometry, RT-PCR, and immunofluorescence staining up to 4 weeks postoperatively. To further elucidate the underlying molecular mechanisms, changes in long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs) were systematically investigated using whole transcriptome sequencing. We then constructed protein-protein interaction networks to find 10 top ranked hub genes among differentially expressed mRNAs. Second, in order to explore the effectiveness of BMSCs and ADSCs on neural tissue-derived ECM materials for repairing PNI, sixty-eight rats were randomized into four groups: ANA group, ANA+BMSC group, ANA+ADSC group, and AUTO group. In the ANA+BMSC and ANA+ADSC groups, ADSCs/BMSCs were equally injected along the long axis of the 10-mm ANA. Then, we performed histological and functional assessments up to 12 weeks postoperatively. RESULTS The results of flow cytometry and RT-PCR showed that ANA combined with BMSCs exhibited more significant immunomodulatory effects, as evidenced by the up-regulation of interleukin (IL)-10, down-regulation of IL-1β and tumor necrosis factor-alpha (TNF-α) expression, promotion of M1-type macrophage polarization to M2-type, and a significant increase in the number of regulatory T cells (Tregs). ANA combined with ADSCs exhibited more pronounced features of pro-myelination and angiogenesis, as evidenced by the up-regulation of myelin-associated protein gene (MBP and MPZ) and angiogenesis-related factors (TGF-β, VEGF). Moreover, differentially expressed genes from whole transcriptome sequencing results further indicated that ANA loaded with BMSCs exhibited notable immunomodulatory effects and ANA loaded with ADSCs was more associated with angiogenesis, axonal growth, and myelin formation. Notably, ANA infused with BMSCs or ADSCs enhanced peripheral nerve regeneration and motor function recovery with no statistically significant differences. CONCLUSIONS This study revealed that both ANA combined with BMSCs and ADSCs enhance peripheral nerve regeneration and motor function recovery, but their biological characteristics (mainly including immunomodulatory effects, pro-vascular regenerative effects, and pro-myelin regenerative effects) and underlying molecular mechanisms in the process of repairing PNI in vivo are different, providing new insights into MSC therapy for peripheral nerve injury and its clinical translation.
Collapse
Affiliation(s)
- Xiangling Li
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Hang Xu
- Department of General Surgery, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Chaochao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yanjun Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yuli Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Tieyuan Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Haofeng Cheng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiangyu Song
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Hebei North University, Zhangjiakou, 075132, China
| | - Zhibo Jia
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Hebei North University, Zhangjiakou, 075132, China
| | - Ruichao He
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jinjuan Zhao
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Shengfeng Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Congcong Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shi Yan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jinpeng Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yu Wei
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jian Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jinshu Tang
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
| | - Jiang Peng
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China.
| | - Yu Wang
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
8
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [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: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
9
|
Wei C, Guo Y, Ci Z, Li M, Zhang Y, Zhou Y. Advances of Schwann cells in peripheral nerve regeneration: From mechanism to cell therapy. Biomed Pharmacother 2024; 175:116645. [PMID: 38729050 DOI: 10.1016/j.biopha.2024.116645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Peripheral nerve injuries (PNIs) frequently occur due to various factors, including mechanical trauma such as accidents or tool-related incidents, as well as complications arising from diseases like tumor resection. These injuries frequently result in persistent numbness, impaired motor and sensory functions, neuropathic pain, or even paralysis, which can impose a significant financial burden on patients due to outcomes that often fall short of expectations. The most frequently employed clinical treatment for PNIs involves either direct sutures of the severed ends or bridging the proximal and distal stumps using autologous nerve grafts. However, autologous nerve transplantation may result in sensory and motor functional loss at the donor site, as well as neuroma formation and scarring. Transplantation of Schwann cells/Schwann cell-like cells has emerged as a promising cellular therapy to reconstruct the microenvironment and facilitate peripheral nerve regeneration. In this review, we summarize the role of Schwann cells and recent advances in Schwann cell therapy in peripheral nerve regeneration. We summarize current techniques used in cell therapy, including cell injection, 3D-printed scaffolds for cell delivery, cell encapsulation techniques, as well as the cell types employed in experiments, experimental models, and research findings. At the end of the paper, we summarize the challenges and advantages of various cells (including ESCs, iPSCs, and BMSCs) in clinical cell therapy. Our goal is to provide the theoretical and experimental basis for future treatments targeting peripheral nerves, highlighting the potential of cell therapy and tissue engineering as invaluable resources for promoting nerve regeneration.
Collapse
Affiliation(s)
- Chuqiao Wei
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuanxin Guo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhen Ci
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Mucong Li
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China.
| |
Collapse
|
10
|
Liu B, Alimi OA, Wang Y, Kong Y, Kuss M, Krishnan MA, Hu G, Xiao Y, Dong J, DiMaio DJ, Duan B. Differentiated mesenchymal stem cells-derived exosomes immobilized in decellularized sciatic nerve hydrogels for peripheral nerve repair. J Control Release 2024; 368:24-41. [PMID: 38367864 PMCID: PMC11411504 DOI: 10.1016/j.jconrel.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Peripheral nerve injury (PNI) and the limitations of current treatments often result in incomplete sensory and motor function recovery, which significantly impact the patient's quality of life. While exosomes (Exo) derived from stem cells and Schwann cells have shown promise on promoting PNI repair following systemic administration or intraneural injection, achieving effective local and sustained Exo delivery holds promise to treat local PNI and remains challenging. In this study, we developed Exo-loaded decellularized porcine nerve hydrogels (DNH) for PNI repair. We successfully isolated Exo from differentiated human adipose-derived mesenchymal stem cells (hADMSC) with a Schwann cell-like phenotype (denoted as dExo). These dExo were further combined with polyethylenimine (PEI), and DNH to create polyplex hydrogels (dExo-loaded pDNH). At a PEI content of 0.1%, pDNH showed cytocompatibility for hADMSCs and supported neurite outgrowth of dorsal root ganglions. The sustained release of dExos from dExo-loaded pDNH persisted for at least 21 days both in vitro and in vivo. When applied around injured nerves in a mouse sciatic nerve crush injury model, the dExo-loaded pDNH group significantly improved sensory and motor function recovery and enhanced remyelination compared to dExo and pDNH only groups, highlighting the synergistic regenerative effects. Interestingly, we observed a negative correlation between the number of colony-stimulating factor-1 receptor (CSF-1R) positive cells and the extent of PNI regeneration at the 21-day post-surgery stage. Subsequent in vitro experiments demonstrated the potential involvement of the CSF-1/CSF-1R axis in Schwann cells and macrophage interaction, with dExo effectively downregulating CSF-1/CSF-1R signaling.
Collapse
Affiliation(s)
- Bo Liu
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Olawale A Alimi
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yanfei Wang
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mena Asha Krishnan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| |
Collapse
|
11
|
Zhang Y, Yi D, Hong Q, Cao J, Geng X, Liu J, Xu C, Cao M, Chen C, Xu S, Zhang Z, Li M, Zhu Y, Peng N. Platelet-rich plasma-derived exosomes boost mesenchymal stem cells to promote peripheral nerve regeneration. J Control Release 2024; 367:265-282. [PMID: 38253204 DOI: 10.1016/j.jconrel.2024.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Peripheral nerve injury (PNI) remains a severe clinical problem with debilitating consequences. Mesenchymal stem cell (MSC)-based therapy is promising, but the problems of poor engraftment and insufficient neurotrophic effects need to be overcome. Herein, we isolated platelet-rich plasma-derived exosomes (PRP-Exos), which contain abundant bioactive molecules, and investigated their potential to increase the regenerative capacity of MSCs. We observed that PRP-Exos significantly increased MSC proliferation, viability, and mobility, decreased MSC apoptosis under stress, maintained MSC stemness, and attenuated MSC senescence. In vivo, PRP-Exo-treated MSCs (pExo-MSCs) exhibited an increased retention rate and heightened therapeutic efficacy, as indicated by increased axonal regeneration, remyelination, and recovery of neurological function in a PNI model. In vitro, pExo-MSCs coculture promoted Schwann cell proliferation and dorsal root ganglion axon growth. Moreover, the increased neurotrophic behaviour of pExo-MSCs was mediated by trophic factors, particularly glia-derived neurotrophic factor (GDNF), and PRP-Exos activated the PI3K/Akt signalling pathway in MSCs, leading to the observed phenotypes. These findings demonstrate that PRP-Exos may be novel agents for increasing the ability of MSCs to promote neural repair and regeneration in patients with PNI.
Collapse
Affiliation(s)
- Yongyi Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China; State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; No.962 Hospital of the PLA Joint Logistic Support Force, Harbin 150080, China
| | - Dan Yi
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Quan Hong
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jiangbei Cao
- Departments of Anaesthesiology, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaodong Geng
- State Key Laboratory of Kidney Diseases, Nephrology Institute of the Chinese PLA, National Clinical Research Centre for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jinwei Liu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chuang Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengyu Cao
- Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Chen
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuaixuan Xu
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhen Zhang
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Molin Li
- Medical School of Chinese PLA, Beijing 100853, China; Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yaqiong Zhu
- Departments of Ultrasound, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| | - Nan Peng
- Medical School of Chinese PLA, Beijing 100853, China; Department of Rehabilitation Medicine, The Second Medical Centre & National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
12
|
Kaviarasan V, Deka D, Balaji D, Pathak S, Banerjee A. Signaling Pathways in Trans-differentiation of Mesenchymal Stem Cells: Recent Advances. Methods Mol Biol 2024; 2736:207-223. [PMID: 37140811 DOI: 10.1007/7651_2023_478] [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] [Indexed: 05/05/2023]
Abstract
Mesenchymal stem cells are a group of multipotent cells that can be induced to differentiate into other cell types. The cells fate is decided by various signaling pathways, growth factors, and transcription factors in differentiation. The proper coordination of these factors will result in cell specification. MSCs are capable of being differentiated into osteogenic, chondrogenic, and adipogenic lineages. Different conditions induces the MSCs into particular phenotypes. The MSC trans-differentiation ensues as a response to environmental factors or due to circumstances that prove to favor trans-differentiation. Depending on the stage at which they are expressed, and the genetic alterations they undergo prior to their expression, transcription factors can accelerate the process of trans-differentiation. Further research has been conducted on the challenging aspect of MSCs being developed into non-mesenchymal lineage. The cells that are differentiated in this way maintain their stability even after being induced in animals. The recent advancements in the trans-differentiation capacities of MSCs on induction with chemicals, growth inducers, improved differentiation mediums, growth factors from plant extracts, and electrical stimulation are discussed in this paper. Signaling pathways have a great effect on MSCs trans-differentiation and they need to be better understood for their applications in therapeutic techniques. So, this paper tends to review the major signaling pathways that play a vital role in the trans-differentiation of MSC.
Collapse
Affiliation(s)
- Vaishak Kaviarasan
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Dikshita Deka
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Darshini Balaji
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India.
| |
Collapse
|
13
|
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
|
14
|
Li J, Wu C, Zeng M, Zhang Y, Wei D, Sun J, Fan H. Functional material-mediated wireless physical stimulation for neuro-modulation and regeneration. J Mater Chem B 2023; 11:9056-9083. [PMID: 37649427 DOI: 10.1039/d3tb01354e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Nerve injuries and neurological diseases remain intractable clinical challenges. Despite the advantages of stem cell therapy in treating neurological disorders, uncontrollable cell fates and loss of cell function in vivo are still challenging. Recently, increasing attention has been given to the roles of external physical signals, such as electricity and ultrasound, in regulating stem cell fate as well as activating or inhibiting neuronal activity, which provides new insights for the treatment of neurological disorders. However, direct physical stimulations in vivo are short in accuracy and safety. Functional materials that can absorb energy from a specific physical field exerted in a wireless way and then release another localized physical signal hold great advantages in mediating noninvasive or minimally invasive accurate indirect physical stimulations to promote the therapeutic effect on neurological disorders. In this review, the mechanism by which various physical signals regulate stem cell fate and neuronal activity is summarized. Based on these concepts, the approaches of using functional materials to mediate indirect wireless physical stimulation for neuro-modulation and regeneration are systematically reviewed. We expect that this review will contribute to developing wireless platforms for neural stimulation as an assistance for the treatment of neurological diseases and injuries.
Collapse
Affiliation(s)
- Jialu Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, Sichuan, China
| | - Mingze Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
15
|
Babu S, Krishnan M, Panneerselvam A, Chinnaiyan M. A comprehensive review on therapeutic application of mesenchymal stem cells in neuroregeneration. Life Sci 2023:121785. [PMID: 37196856 DOI: 10.1016/j.lfs.2023.121785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Each year, thousands of people suffer from traumatic peripheral nerve lesions, which impair mobility and sensibility and frequently have fatal outcomes. The recovery of peripheral nerves on its own is frequently insufficient. In this regard, cell therapy is currently one of the most cutting-edge techniques for nerve healing. The purpose of this review is to highlight the properties of various types of mesenchymal stem cells (MSCs) that are critical for peripheral nerve regeneration after nerve injury. The Preferred Reporting term used to review the available literature are "nerve regeneration," "stem cells," "peripheral nerve damage," "rat," and "human" were combined. In addition, using the phrases "stem cells" and "nerve regeneration" in PubMed, a "MeSH" search was conducted. This study describes the features of the most often utilized MSCs, as well as its paracrine potential, targeted stimulation, and propensity for differentiation into Schwann-like and neuronal-like cells. For the repair of peripheral nerve lesions, ADSCs appear to be the most relevant and promising MSCs, because of their ability to sustain and increase axonal growth, as well as their outstanding paracrine activity, putative differentiation potential, low immunogenicity, and excellent post-transplant survival rate.
Collapse
Affiliation(s)
- Shyamaladevi Babu
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India.
| | - Madhan Krishnan
- Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India
| | | | - Mayilvanan Chinnaiyan
- Department of Otolaryngology, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| |
Collapse
|
16
|
Mao S, Fonder C, Rubby MF, Phillips GJ, Sakaguchi DS, Que L. An integrated microfluidic chip for studying the effects of neurotransmitters on neurospheroids. LAB ON A CHIP 2023; 23:1649-1663. [PMID: 36751868 DOI: 10.1039/d2lc00755j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
To improve our understanding of how the central nervous system functions in health and disease, we report the development of an integrated chip for studying the effects of the neurotransmitters dopamine and serotonin on adult rat hippocampal progenitor cell (AHPC) neurospheroids. This chip allows dopamine or serotonin located in one chamber to diffuse to AHPC neurospheroids cultured in an adjacent chamber through a built-in diffusion barrier created by an array of intentionally misaligned micropillars. The gaps among the micropillars are filled with porous poly(ethylene glycol) (PEG) gel to tune the permeability of the diffusion barrier. An electrochemical sensor is also integrated within the chamber where the neurospheroids can be cultured, thereby allowing monitoring of the concentrations of dopamine or serotonin. Experiments show that concentrations of the neurotransmitters inside the neurospheroid chamber can be increased over a period of several hours to over 10 days by controlling the compositions of the PEG gel inside the diffusion barrier. The AHPC neurospheroids cultured in the chip remain highly viable following dopamine or serotonin treatment. Cell proliferation and neuronal differentiation have also been observed following treatment, revealing that the AHPC neurospheroids are a valuable in vitro brain model for neurogenesis research. Finally, we show that by tuning the permeability of diffusion barrier, we can block transfer of Escherichia coli cells across the diffusion barrier, while allowing dopamine or serotonin to pass through. These results suggest the feasibility of using the chip to better understand the interactions between microbiota and brain via the gut-brain axis.
Collapse
Affiliation(s)
- Subin Mao
- Department of Electrical and Computer Engineering, Iowa State University, Ames IA 50011, USA.
| | - Catherine Fonder
- Molecular, Cellular, and Developmental Biology Program, Iowa State University, Ames IA 50011, USA.
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames IA 50011, USA
| | - Md Fazlay Rubby
- Department of Electrical and Computer Engineering, Iowa State University, Ames IA 50011, USA.
| | - Gregory J Phillips
- Department of Veterinary Microbiology, Iowa State University, Ames IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames IA 50011, USA
| | - Donald S Sakaguchi
- Molecular, Cellular, and Developmental Biology Program, Iowa State University, Ames IA 50011, USA.
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames IA 50011, USA
- Neuroscience Program, Iowa State University, Ames IA 50011, USA
- Nanovaccine Institute, Iowa State University, Ames IA 50011, USA
| | - Long Que
- Department of Electrical and Computer Engineering, Iowa State University, Ames IA 50011, USA.
| |
Collapse
|
17
|
Sharifi M, Farahani MK, Salehi M, Atashi A, Alizadeh M, Kheradmandi R, Molzemi S. Exploring the Physicochemical, Electroactive, and Biodelivery Properties of Metal Nanoparticles on Peripheral Nerve Regeneration. ACS Biomater Sci Eng 2023; 9:106-138. [PMID: 36545927 DOI: 10.1021/acsbiomaterials.2c01216] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite the advances in the regeneration/rehabilitation field of damaged tissues, the functional recovery of peripheral nerves (PNs), especially in a long gap injury, is considered a great medical challenge. Recent progress in nanomedicine has provided great hope for PN regeneration through the strategy of controlling cell behavior by metal nanoparticles individually or loaded on scaffolds/conduits. Despite the confirmed toxicity of metal nanoparticles due to long-term accumulation in nontarget tissues, they play a role in the damaged PN regeneration based on the topography modification of scaffolds/conduits, enhancing neurotrophic factor secretion, the ion flow improvement, and the regulation of electrical signals. Determining the fate of neural progenitor cells would be a major achievement in PN regeneration, which seems to be achievable by metal nanoparticles through altering cell vital approaches and controlling their functions. Therefore, in this literature, an attempt was made to provide an overview of the effective activities of metal nanoparticles on the PN regeneration, until the vital clues of the PN regeneration and how they are changed by metal nanoparticles are revealed to the researcher.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran.,Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| | - Mohammad Kamalabadi Farahani
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran.,Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Faculty of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| | - Rasoul Kheradmandi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran.,Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| | - Sahar Molzemi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran.,Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773955, Iran
| |
Collapse
|
18
|
Li X, Zhang X, Hao M, Wang D, Jiang Z, Sun L, Gao Y, Jin Y, Lei P, Zhuo Y. The application of collagen in the repair of peripheral nerve defect. Front Bioeng Biotechnol 2022; 10:973301. [PMID: 36213073 PMCID: PMC9542778 DOI: 10.3389/fbioe.2022.973301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Collagen is a natural polymer expressed in the extracellular matrix of the peripheral nervous system. It has become increasingly crucial in peripheral nerve reconstruction as it was involved in regulating Schwann cell behaviors, maintaining peripheral nerve functions during peripheral nerve development, and being strongly upregulated after nerve injury to promote peripheral nerve regeneration. Moreover, its biological properties, such as low immunogenicity, excellent biocompatibility, and biodegradability make it a suitable biomaterial for peripheral nerve repair. Collagen provides a suitable microenvironment to support Schwann cells’ growth, proliferation, and migration, thereby improving the regeneration and functional recovery of peripheral nerves. This review aims to summarize the characteristics of collagen as a biomaterial, analyze its role in peripheral nerve regeneration, and provide a detailed overview of the recent advances concerning the optimization of collagen nerve conduits in terms of physical properties and structure, as well as the application of the combination with the bioactive component in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Xiaolan Li
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Hao
- School of Acupuncture-Moxi Bustion and Tuina, Changchun University of Chinese Medicine, Changchun, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Ziping Jiang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, China
| | - Liqun Sun
- Department of Pediatrics, First Hospital of Jilin University, Changchun, China
| | - Yongjian Gao
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ye Jin
- Department of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Peng Lei, ; Yue Zhuo,
| | - Yue Zhuo
- School of Acupuncture-Moxi Bustion and Tuina, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Peng Lei, ; Yue Zhuo,
| |
Collapse
|
19
|
Liu B, Kong Y, Shi W, Kuss M, Liao K, Hu G, Xiao P, Sankarasubramanian J, Guda C, Wang X, Lei Y, Duan B. Exosomes derived from differentiated human ADMSC with the Schwann cell phenotype modulate peripheral nerve-related cellular functions. Bioact Mater 2022; 14:61-75. [PMID: 35310346 PMCID: PMC8892082 DOI: 10.1016/j.bioactmat.2021.11.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral nerve regeneration remains a significant clinical challenge due to the unsatisfactory functional recovery and public health burden. Exosomes, especially those derived from mesenchymal stem cells (MSCs), are promising as potential cell-free therapeutics and gene therapy vehicles for promoting neural regeneration. In this study, we reported the differentiation of human adipose derived MSCs (hADMSCs) towards the Schwann cell (SC) phenotype (hADMSC-SCs) and then isolated exosomes from hADMSCs with and without differentiation (i.e., dExo vs uExo). We assessed and compared the effects of uExo and dExo on antioxidative, angiogenic, anti-inflammatory, and axon growth promoting properties by using various peripheral nerve-related cells. Our results demonstrated that hADMSC-SCs secreted more neurotrophic factors and other growth factors, compared to hADMSCs without differentiation. The dExo isolated from hADMSC-SCs protected rat SCs from oxidative stress and enhanced HUVEC migration and angiogenesis. Compared to uExo, dExo also had improved performances in downregulating pro-inflammatory gene expressions and cytokine secretions and promoting axonal growth of sensory neurons differentiated from human induced pluripotent stem cells. Furthermore, microRNA (miRNA) sequencing analysis revealed that exosomes and their parent cells shared some similarities in their miRNA profiles and exosomes displayed a distinct miRNA signature. Many more miRNAs were identified in dExo than in uExo. Several upregulated miRNAs, like miRNA-132-3p and miRNA-199b-5p, were highly related to neuroprotection, anti-inflammation, and angiogenesis. The dExo can effectively modulate various peripheral nerve-related cellular functions and is promising for cell-free biological therapeutics to enhance neural regeneration. Exosomes were isolated from hADMSCs with and without differentiation towards SC phenotype (i.e., dExo vs uExo). hADMSC-SCs secreted more growth factors compared to hADMSCs without differentiation. The dExo protected rat SCs from oxidative stress and enhanced endothelial cell migration and angiogenesis. dExo promoted axonal growth of sensory neurons differentiated from hiPSCs. miRNA sequencing analysis unveiled and compared the exosomal and cellular miRNA profiles.
Collapse
|
20
|
Borah R, Das JM, Upadhyay J. Surface Functionalized Polyaniline Nanofibers:Chitosan Nanocomposite for Promoting Neuronal-like Differentiation of Primary Adipose Derived Mesenchymal Stem Cells and Urease Activity. ACS APPLIED BIO MATERIALS 2022; 5:3193-3211. [PMID: 35775198 DOI: 10.1021/acsabm.2c00171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bioscaffolds having electrically conducting polymers (CPs) have become increasingly relevant in tissue engineering (TE) because of their ability to regulate conductivity and promote biological function. With this in mind, the current study shows a conducting polyaniline nanofibers (PNFs) dispersed chitosan (Ch) nanocomposites scaffold with a simple one-step surface functionalization approach using glutaraldehyde for potential neural regeneration applications. According to the findings, 4 wt % PNFs dispersion in Ch matrix is an optimal concentration for achieving desirable biological functions while maintaining required physicochemical properties as evidenced by SEM, XRD, current-voltage (I-V) measurement, mechanical strength test, and in vitro biodegradability test. Surface chemical compositional analysis using XPS and ATR FT-IR confirms the incorporation of aldehyde functionality after functionalization, which is corroborated by surface energy calculations following the Van Oss-Chaudhury-Good method. Surface functionalization induced enhancement in surface hydrophilicity in terms of the polar component of surface energy (γiAB) from 6.35 to 12.54 mN m-1 along with an increase in surface polarity from 13.61 to 22.54%. Functionalized PNF:Ch scaffolds demonstrated improvement in enzyme activity from 67 to 94% and better enzyme kinetics with a reduction of Michaelis constants (Km) from 21.55 to 13.81 mM, indicating favorable protein-biomaterial interactions and establishing them as biologically perceptible materials. Surface functionalization mediated improved cell-biomaterial interactions led to improved viability, adhesion, and spreading of primary adipose derived mesenchymal stem cells (ADMSCs) as well as improved immunocompatibility. Cytoskeletal architecture assessment under differentiating media containing 10 ng/mL of each basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) revealed significant actin remodeling with neurite-like projections on the functionalized scaffolds after 14 days. Immunocytochemistry results showed that more than 85% of cells expressed early neuron specific β III tubulin protein on the functionalized scaffolds, whereas glial fibrillary acidic protein (GFAP) expression was limited to approximately 40% of cells. The findings point to the functionalized nanocomposites' potential as a smart scaffold for electrically stimulated neural regeneration, as they are flexible enough to be designed into microchanneled or conduit-like structures that mimic the microstructures and mechanical properties of peripheral nerves.
Collapse
Affiliation(s)
- Rajiv Borah
- Seri-Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati 781035, India
| | - Jitu Mani Das
- Seri-Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science & Technology, Guwahati 781035, India
| | - Jnanendra Upadhyay
- Department of Physics, Dakshin Kamrup College, Kamrup, Assam 781125, India
| |
Collapse
|
21
|
Tang N, Wang X, Zhu J, Sun K, Li S, Tao K. Labelling stem cells with a nanoprobe for evaluating the homing behaviour in facial nerve injury repair. Biomater Sci 2022; 10:808-818. [PMID: 34989358 DOI: 10.1039/d1bm01823j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
It is crucial and clinically relevant to clarify the homing efficiency and retention of stem cells in different implanting strategies of cell therapy for various injuries. However, the need for a tool for investigating the mechanisms is still unmet. We herein introduce multi-modal BaGdF5:Yb,Tm nanoparticles as a nanoprobe to label adipose-derived stem cells (ADSCs) and detect the homing behavior with a micro-computed tomography (micro-CT) imaging technique. The migration of cells injected locally or intravenously, with or without a chemokine, CXCL 12, was compared. A higher homing efficiency of ADSCs was observed in both intravenously injected groups, in contrast to the low efficiency of cell retention in local implantation. Meanwhile, CXCL 12 promoted the homing of ADSCs, especially in the intravenous route. Nonetheless, the administration of CXCL 12 showed its therapeutic efficacy, whereas intravenous injection of ADSCs almost did not. Our work provided a tool for in vivo imaging of the behavior of implanted cells in preclinical studies of cell therapy, and more importantly, implied that the parameters for implanting stem cells in clinical operation should be carefully considered.
Collapse
Affiliation(s)
- Na Tang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Xueyi Wang
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China.
| | - Jin Zhu
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China.
| | - Kang Sun
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Shiting Li
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P. R. China.
| | - Ke Tao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| |
Collapse
|
22
|
Halim A, Qu KY, Zhang XF, Huang NP. Recent Advances in the Application of Two-Dimensional Nanomaterials for Neural Tissue Engineering and Regeneration. ACS Biomater Sci Eng 2021; 7:3503-3529. [PMID: 34291638 DOI: 10.1021/acsbiomaterials.1c00490] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complexity of the nervous system structure and function, and its slow regeneration rate, makes it more difficult to treat compared to other tissues in the human body when an injury occurs. Moreover, the current therapeutic approaches including the use of autografts, allografts, and pharmacological agents have several drawbacks and can not fully restore nervous system injuries. Recently, nanotechnology and tissue engineering approaches have attracted many researchers to guide tissue regeneration in an effective manner. Owing to their remarkable physicochemical and biological properties, two-dimensional (2D) nanomaterials have been extensively studied in the tissue engineering and regenerative medicine field. The great conductivity of these materials makes them a promising candidate for the development of novel scaffolds for neural tissue engineering application. Moreover, the high loading capacity of 2D nanomaterials also has attracted many researchers to utilize them as a drug/gene delivery method to treat various devastating nervous system disorders. This review will first introduce the fundamental physicochemical properties of 2D nanomaterials used in biomedicine and the supporting biological properties of 2D nanomaterials for inducing neuroregeneration, including their biocompatibility on neural cells, the ability to promote the neural differentiation of stem cells, and their immunomodulatory properties which are beneficial for alleviating chronic inflammation at the site of the nervous system injury. It also discusses various types of 2D nanomaterials-based scaffolds for neural tissue engineering applications. Then, the latest progress on the use of 2D nanomaterials for nervous system disorder treatment is summarized. Finally, a discussion of the challenges and prospects of 2D nanomaterials-based applications in neural tissue engineering is provided.
Collapse
Affiliation(s)
- Alexander Halim
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Kai-Yun Qu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Xiao-Feng Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
23
|
Lavorato A, Raimondo S, Boido M, Muratori L, Durante G, Cofano F, Vincitorio F, Petrone S, Titolo P, Tartara F, Vercelli A, Garbossa D. Mesenchymal Stem Cell Treatment Perspectives in Peripheral Nerve Regeneration: Systematic Review. Int J Mol Sci 2021; 22:E572. [PMID: 33430035 PMCID: PMC7827385 DOI: 10.3390/ijms22020572] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic peripheral nerve lesions affect hundreds of thousands of patients every year; their consequences are life-altering and often devastating and cause alterations in movement and sensitivity. Spontaneous peripheral nerve recovery is often inadequate. In this context, nowadays, cell therapy represents one of the most innovative approaches in the field of nerve repair therapies. The purpose of this systematic review is to discuss the features of different types of mesenchymal stem cells (MSCs) relevant for peripheral nerve regeneration after nerve injury. The published literature was reviewed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A combination of the keywords "nerve regeneration", "stem cells", "peripheral nerve injury", "rat", and "human" were used. Additionally, a "MeSH" research was performed in PubMed using the terms "stem cells" and "nerve regeneration". The characteristics of the most widely used MSCs, their paracrine potential, targeted stimulation, and differentiation potentials into Schwann-like and neuronal-like cells are described in this paper. Considering their ability to support and stimulate axonal growth, their remarkable paracrine activity, their presumed differentiation potential, their extremely low immunogenicity, and their high survival rate after transplantation, ADSCs appear to be the most suitable and promising MSCs for the recovery of peripheral nerve lesion. Clinical considerations are finally reported.
Collapse
Affiliation(s)
- Andrea Lavorato
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (S.R.); (L.M.)
| | - Marina Boido
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (M.B.); (A.V.)
| | - Luisa Muratori
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (S.R.); (L.M.)
| | - Giorgia Durante
- Faculty of Medicine and Surgery, University of Turin, 10126 Turin, TO, Italy;
| | - Fabio Cofano
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Francesca Vincitorio
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Salvatore Petrone
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| | - Paolo Titolo
- Traumatology–Reconstructive Microsurgery, Department of Orthopaedics and Traumatology, CTO Hospital, 10126 Turin, TO, Italy;
| | - Fulvio Tartara
- Neurosurgery Unit, Istituto Clinico Città Studi (ICCS), 20131 Milan, MI, Italy;
| | - Alessandro Vercelli
- Department of Neuroscience “Rita Levi Montalcini”, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, TO, Italy; (M.B.); (A.V.)
| | - Diego Garbossa
- Neurosurgery Unit, Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, TO, Italy; (F.C.); (F.V.); (S.P.); (D.G.)
| |
Collapse
|
24
|
Silencing Celsr2 inhibits the proliferation and migration of Schwann cells through suppressing the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2020; 533:623-630. [PMID: 32988580 DOI: 10.1016/j.bbrc.2020.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 01/26/2023]
Abstract
After a peripheral nerve injury, the remaining Schwann cells undergo proliferation and adopt a migratory phenotype to prepare for the regeneration of nerves. Celsr2 has been reported to play an important role in the development and maintenance of the function of the nervous system. However, the role and mechanism of Celsr2 during peripheral nerve regeneration remain unknown. Here, we showed that after sciatic nerve injury, Celsr2 mRNA and protein were significantly increased in nerve tissues. In addition, silencing Celsr2 decreased the ki67-positve portion and the migration distance of Schwann cells in vivo. In vitro, the results of MTT and EdU staining, transwell and wound healing assays indicated that Celsr2 siRNA-transfected primary Schwann cells showed significant decrease in proliferation and migration compared to that seen in negative control (NC)-transfected cells. Furthermore, we found that Wnt/β-catenin luciferase activity was reduced, as were the expression of β-catenin in the nucleus and the mRNA levels of its downstream genes Cyclin D1 and MMP-7 in Celsr2 siRNA-transfected primary Schwann cells. Further investigations showed that silencing Celsr2 inhibited the phosphorylation of GSK3β. Moreover, specific activators of the Wnt/β-catenin pathway, LiCl or mutant β-catenin (S33Y), partially reversed the inhibitory effect of Celsr2 siRNA. Taken together, our data indicated that silencing Celsr2 inhibited Schwann cells migration and proliferation through the suppressing Wnt/β-catenin pathway, providing a potential target for peripheral nerve regeneration.
Collapse
|
25
|
Keshavarz M, Wales DJ, Seichepine F, Abdelaziz MEMK, Kassanos P, Li Q, Temelkuran B, Shen H, Yang GZ. Induced neural stem cell differentiation on a drawn fiber scaffold-toward peripheral nerve regeneration. ACTA ACUST UNITED AC 2020; 15:055011. [PMID: 32330920 DOI: 10.1088/1748-605x/ab8d12] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
To achieve regeneration of long sections of damaged nerves, restoration methods such as direct suturing or autologous grafting can be inefficient. Solutions involving biohybrid implants, where neural stem cells are grown in vitro on an active support before implantation, have attracted attention. Using such an approach, combined with recent advancements in microfabrication technology, the chemical and physical environment of cells can be tailored in order to control their behaviors. Herein, a neural stem cell polycarbonate fiber scaffold, fabricated by 3D printing and thermal drawing, is presented. The combined effect of surface microstructure and chemical functionalization using poly-L-ornithine (PLO) and double-walled carbon nanotubes (DWCNTs) on the biocompatibility of the scaffold, induced differentiation of the neural stem cells (NSCs) and channeling of the neural cells was investigated. Upon treatment of the fiber scaffold with a suspension of DWCNTs in PLO (0.039 g l-1) and without recombinants a high degree of differentiation of NSCs into neuronal cells was confirmed by using nestin, galactocerebroside and doublecortin immunoassays. These findings illuminate the potential use of this biohybrid approach for the realization of future nerve regenerative implants.
Collapse
Affiliation(s)
- Meysam Keshavarz
- Hamlyn Centre for Robotic Surgery, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yao B, Wang R, Wang Y, Zhang Y, Hu T, Song W, Li Z, Huang S, Fu X. Biochemical and structural cues of 3D-printed matrix synergistically direct MSC differentiation for functional sweat gland regeneration. SCIENCE ADVANCES 2020; 6:eaaz1094. [PMID: 32181358 PMCID: PMC7056319 DOI: 10.1126/sciadv.aaz1094] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/10/2019] [Indexed: 05/10/2023]
Abstract
Mesenchymal stem cells (MSCs) encapsulation by three-dimensionally (3D) printed matrices were believed to provide a biomimetic microenvironment to drive differentiation into tissue-specific progeny, which made them a great therapeutic potential for regenerative medicine. Despite this potential, the underlying mechanisms of controlling cell fate in 3D microenvironments remained relatively unexplored. Here, we bioprinted a sweat gland (SG)-like matrix to direct the conversion of MSC into functional SGs and facilitated SGs recovery in mice. By extracellular matrix differential protein expression analysis, we identified that CTHRC1 was a critical biochemical regulator for SG specification. Our findings showed that Hmox1 could respond to the 3D structure activation and also be involved in MSC differentiation. Using inhibition and activation assay, CTHRC1 and Hmox1 synergistically boosted SG gene expression profile. Together, these findings indicated that biochemical and structural cues served as two critical impacts of 3D-printed matrix on MSC fate decision into the glandular lineage and functional SG recovery.
Collapse
Affiliation(s)
- Bin Yao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, P. R. China
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Rui Wang
- Chinese PLA 306 Hospital, Beijing 100000, P.R. China
| | - Yihui Wang
- Handan People’s Hospital, Hebei 056000, P.R. China
| | - Yijie Zhang
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, P. R. China
| | - Tian Hu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, P. R. China
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Wei Song
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
| | - Zhao Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, P. R. China
| | - Sha Huang
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, P. R. China
- Corresponding author. (S.H.); (X.F.)
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA, Beijing 100853, P. R. China
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, First Affiliated Hospital of PLA General Hospital, Beijing 100048, P.R. China
- Corresponding author. (S.H.); (X.F.)
| |
Collapse
|
27
|
Jang SR, Kim JI, Park CH, Kim CS. The controlled design of electrospun PCL/silk/quercetin fibrous tubular scaffold using a modified wound coil collector and L-shaped ground design for neural repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110776. [PMID: 32279813 DOI: 10.1016/j.msec.2020.110776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 12/28/2022]
Abstract
Asymmetrically porous and aligned fibrous tubular conduit with selective permeability as a biomimetic neural scaffold was manufactured using polycaprolactone (PCL), silk, and quercetin by a modified electrospinning method. The outer surface of the randomly oriented fibrous scaffold had microscale pores that could prevent fibrous tissue invasion (FTI), but could permeate neurotrophic factors, nutrients, and oxygen. The inner surface of the aligned fibrous scaffold can be favorable for neurite outgrowth, because of their superior neural cell attachment, migration, and directional growth. In vitro and in vivo studies have demonstrated the therapeutic effect of Quercetin, a ubiquitous flavonoid widely distributed in plants, on neuropathy, by modulating the expression of NRF-2-dependent antioxidant responsive elements. In this study, the controlled inner and outer surface geometry of the 0.5, 1.0, and 2.0 wt% quercetin-containing electrospun PCL/silk fibrous tubular scaffold fabricated via a modified wound coil collector and L-shaped ground design (WCC-LG) was characterized by FE-SEM, TEM, FFT, FT-IR, and XRD. In addition, two types of neural cell lines, PC12 and S42, were used to evaluate the cell proliferation rate of the different amount of quercetin-loaded PCL/silk tubular scaffolds.
Collapse
Affiliation(s)
- Se Rim Jang
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Jeong In Kim
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea
| | - Chan Hee Park
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Division of Mechanical Design Engineering, College of Engineering, Jeonbuk National University, Jeonju 561-756, Republic of Korea.
| | - Cheol Sang Kim
- Department of Bionanosystem Engineering, Graduate School, Jeonbuk National University, Jeonju 561-756, Republic of Korea; Division of Mechanical Design Engineering, College of Engineering, Jeonbuk National University, Jeonju 561-756, Republic of Korea.
| |
Collapse
|
28
|
Luo Y, Yang H, Zhou YF, Hu B. Dual and multi-targeted nanoparticles for site-specific brain drug delivery. J Control Release 2019; 317:195-215. [PMID: 31794799 DOI: 10.1016/j.jconrel.2019.11.037] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/26/2022]
Abstract
In recent years, nanomedicines have emerged as a promising method for central nervous system drug delivery, enabling the drugs to overcome the blood-brain barrier and accumulate preferentially in the brain. Despite the current success of brain-targeted nanomedicines, limitations still exist in terms of the targeting specificity. Based on the molecular mechanism, the exact cell populations and subcellular organelles where the injury occurs and the drugs take effect have been increasingly accepted as a more specific target for the next generation of nanomedicines. Dual and multi-targeted nanoparticles integrate different targeting functionalities and have provided a paradigm for precisely delivering the drug to the pathological site inside the brain. The targeting process often involves the sequential or synchronized navigation of the targeting moieties, which allows highly controlled drug delivery compared to conventional targeting strategies. Herein, we focus on the up-to-date design of pathological site-specific nanoparticles for brain drug delivery, highlighting the dual and multi-targeting strategies that were employed and their impact on improving targeting specificity and therapeutic effects. Furthermore, the background discussion of the basic properties of a brain-targeted nanoparticle and the common lesion features classified by neurological pathology are systematically summarized.
Collapse
Affiliation(s)
- Yan Luo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hang Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-Fan Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
29
|
|
30
|
Determination of Electrical Stimuli Parameters To Transdifferentiate Genetically Engineered Mesenchymal Stem Cells into Neuronal or Glial Lineages. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00126-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
31
|
Kang Y, Liu Y, Liu Z, Ren S, Xiong H, Chen J, Duscher D, Machens HG, Liu W, Guo G, Zhan P, Chen H, Chen Z. Differentiated human adipose-derived stromal cells exhibit the phenotypic and functional characteristics of mature Schwann cells through a modified approach. Cytotherapy 2019; 21:987-1003. [PMID: 31351800 DOI: 10.1016/j.jcyt.2019.04.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/09/2019] [Accepted: 04/29/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS Tissue engineering technology is a promising therapeutic strategy in peripheral nerve injury. Schwann cells (SCs) are deemed to be a vital component of cell-based nerve regeneration therapies. Many methods for producing SC-like cells derived from adipose-derived stromal cells (ADSCs) have been explored, but their phenotypic and functional characteristics remain unsatisfactory. METHODS We investigated whether human ADSCs can be induced to differentiate into mature and stable SC-like cells with the addition of insulin, progestero``ne and glucocorticoids. The phenotypic and functional characteristics of new differentiated ADSCs (modified SC-like cells) were evaluated by real-time quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and immunocytochemistry in vitro. Cells loaded into collagen sponge biomaterials were implanted around transected sciatic nerves with a 10-mm gap in vivo. The axon regrowth and functional recovery of the regenerated nerves were assessed by immunohistochemistry and Walking footprint analysis. RESULTS After differentiation induction, the modified SC-like cells showed significantly up-regulated levels of S100B and P0 and enhanced proliferative and migratory capacities. In addition, the modified SC-like cells showed increased secretion of neurotrophic factors, and their functional characteristics were maintained for more than 3 weeks after removing the induction reagents. The modified SC-like cells exhibited significantly enhanced axon regrowth, myelination and functional recovery after sciatic nerve injury. CONCLUSIONS Overall, the results suggest that this modified induction method can induce human ADSCs to differentiate into cells with the molecular and functional properties of mature SCs and increase the promotion of peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yutian Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenyu Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sen Ren
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hewei Xiong
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Dominik Duscher
- Department of Plastic and Hand Surgery, Klinikum rechts der Isar (MRI), Technische Universität München (TUM), Ismaninger Straße 22 81675, München, Germany
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum rechts der Isar (MRI), Technische Universität München (TUM), Ismaninger Straße 22 81675, München, Germany
| | - Wei Liu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guojun Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongrui Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
32
|
Uz M, Jackson K, Donta MS, Jung J, Lentner MT, Hondred JA, Claussen JC, Mallapragada SK. Fabrication of High-resolution Graphene-based Flexible Electronics via Polymer Casting. Sci Rep 2019; 9:10595. [PMID: 31332270 PMCID: PMC6646327 DOI: 10.1038/s41598-019-46978-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
In this study, a novel method based on the transfer of graphene patterns from a rigid or flexible substrate onto a polymeric film surface via solvent casting was developed. The method involves the creation of predetermined graphene patterns on the substrate, casting a polymer solution, and directly transferring the graphene patterns from the substrate to the surface of the target polymer film via a peeling-off method. The feature sizes of the graphene patterns on the final film can vary from a few micrometers (as low as 5 µm) to few millimeters range. This process, applied at room temperature, eliminates the need for harsh post-processing techniques and enables creation of conductive graphene circuits (sheet resistance: ~0.2 kΩ/sq) with high stability (stable after 100 bending and 24 h washing cycles) on various polymeric flexible substrates. Moreover, this approach allows precise control of the substrate properties such as composition, biodegradability, 3D microstructure, pore size, porosity and mechanical properties using different film formation techniques. This approach can also be used to fabricate flexible biointerfaces to control stem cell behavior, such as differentiation and alignment. Overall, this promising approach provides a facile and low-cost method for the fabrication of flexible and stretchable electronic circuits.
Collapse
Affiliation(s)
- Metin Uz
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - Kyle Jackson
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - Maxsam S Donta
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - Juhyung Jung
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - Matthew T Lentner
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - John A Hondred
- Department of Mechanical Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - Jonathan C Claussen
- Department of Mechanical Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
33
|
Uz M, Donta M, Mededovic M, Sakaguchi DS, Mallapragada SK. Development of Gelatin and Graphene-Based Nerve Regeneration Conduits Using Three-Dimensional (3D) Printing Strategies for Electrical Transdifferentiation of Mesenchymal Stem Cells. Ind Eng Chem Res 2019. [DOI: 10.1021/acs.iecr.8b05537] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Metin Uz
- Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011-2230, United States
| | - Maxsam Donta
- Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011-2230, United States
| | - Meryem Mededovic
- Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011-2230, United States
| | - Donald S. Sakaguchi
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011-1079, United States
| | - Surya K. Mallapragada
- Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011-2230, United States
| |
Collapse
|
34
|
De la Rosa MB, Kozik EM, Sakaguchi DS. Adult Stem Cell-Based Strategies for Peripheral Nerve Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:41-71. [PMID: 30151648 DOI: 10.1007/5584_2018_254] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peripheral nerve injuries (PNI) occur as the result of sudden trauma and can lead to life-long disability, reduced quality of life, and heavy economic and social burdens. Although the peripheral nervous system (PNS) has the intrinsic capacity to regenerate and regrow axons to a certain extent, current treatments frequently show incomplete recovery with poor functional outcomes, particularly for large PNI. Many surgical procedures are available to halt the propagation of nerve damage, and the choice of a procedure depends on the extent of the injury. In particular, recovery from large PNI gaps is difficult to achieve without any therapeutic intervention or some form of tissue/cell-based therapy. Autologous nerve grafting, considered the "gold standard" is often implemented for treatment of gap formation type PNI. Although these surgical procedures provide many benefits, there are still considerable limitations associated with such procedures as donor site morbidity, neuroma formation, fascicle mismatch, and scarring. To overcome such restrictions, researchers have explored various avenues to improve post-surgical outcomes. The most commonly studied methods include: cell transplantation, growth factor delivery to stimulate regenerating axons and implanting nerve guidance conduits containing replacement cells at the site of injury. Replacement cells which offer maximum benefits for the treatment of PNI, are Schwann cells (SCs), which are the peripheral glial cells and in part responsible for clearing out debris from the site of injury. Additionally, they release growth factors to stimulate myelination and axonal regeneration. Both primary SCs and genetically modified SCs enhance nerve regeneration in animal models; however, there is no good source for extracting SCs and the only method to obtain SCs is by sacrificing a healthy nerve. To overcome such challenges, various cell types have been investigated and reported to enhance nerve regeneration.In this review, we have focused on cell-based strategies aimed to enhance peripheral nerve regeneration, in particular the use of mesenchymal stem cells (MSCs). Mesenchymal stem cells are preferred due to benefits such as autologous transplantation, routine isolation procedures, and paracrine and immunomodulatory properties. Mesenchymal stem cells have been transplanted at the site of injury either directly in their native form (undifferentiated) or in a SC-like form (transdifferentiated) and have been shown to significantly enhance nerve regeneration. In addition to transdifferentiated MSCs, some studies have also transplanted ex-vivo genetically modified MSCs that hypersecrete growth factors to improve neuroregeneration.
Collapse
Affiliation(s)
- Metzere Bierlein De la Rosa
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.,Veterinary Specialty Center, Buffalo Grove, IL, USA
| | - Emily M Kozik
- Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA.,Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Donald S Sakaguchi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA. .,Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA. .,Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA. .,Neuroscience Program, Iowa State University, Ames, IA, USA.
| |
Collapse
|