1
|
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
|
2
|
Broeren BO, Hundepool CA, Kumas AH, Duraku LS, Walbeehm ET, Hooijmans CR, Power DM, Zuidam JM, De Jong T. The effectiveness of acellular nerve allografts compared to autografts in animal models: A systematic review and meta-analysis. PLoS One 2024; 19:e0279324. [PMID: 38295088 PMCID: PMC10829984 DOI: 10.1371/journal.pone.0279324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/07/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Treatment of nerve injuries proves to be a worldwide clinical challenge. Acellular nerve allografts are suggested to be a promising alternative for bridging a nerve gap to the current gold standard, an autologous nerve graft. OBJECTIVE To systematically review the efficacy of the acellular nerve allograft, its difference from the gold standard (the nerve autograft) and to discuss its possible indications. MATERIAL AND METHODS PubMed, Embase and Web of Science were systematically searched until the 4th of January 2022. Original peer reviewed paper that presented 1) distinctive data; 2) a clear comparison between not immunologically processed acellular allografts and autologous nerve transfers; 3) was performed in laboratory animals of all species and sex. Meta analyses and subgroup analyses (for graft length and species) were conducted for muscle weight, sciatic function index, ankle angle, nerve conduction velocity, axon count diameter, tetanic contraction and amplitude using a Random effects model. Subgroup analyses were conducted on graft length and species. RESULTS Fifty articles were included in this review and all were included in the meta-analyses. An acellular allograft resulted in a significantly lower muscle weight, sciatic function index, ankle angle, nerve conduction velocity, axon count and smaller diameter, tetanic contraction compared to an autologous nerve graft. No difference was found in amplitude between acellular allografts and autologous nerve transfers. Post hoc subgroup analyses of graft length showed a significant reduced muscle weight in long grafts versus small and medium length grafts. All included studies showed a large variance in methodological design. CONCLUSION Our review shows that the included studies, investigating the use of acellular allografts, showed a large variance in methodological design and are as a consequence difficult to compare. Nevertheless, our results indicate that treating a nerve gap with an allograft results in an inferior nerve recovery compared to an autograft in seven out of eight outcomes assessed in experimental animals. In addition, based on our preliminary post hoc subgroup analyses we suggest that when an allograft is being used an allograft in short and medium (0-1cm, > 1-2cm) nerve gaps is preferred over an allograft in long (> 2cm) nerve gaps.
Collapse
Affiliation(s)
- Berend O. Broeren
- Department of Plastic & Reconstructive Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Caroline A. Hundepool
- Department of Plastic & Reconstructive Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Ali H. Kumas
- Department of Plastic & Reconstructive Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Liron S. Duraku
- Department of Plastic, Reconstructive & Hand Surgery, Amsterdam UMC, Amsterdam, The Netherlands
| | - Erik T. Walbeehm
- Department of Plastic, Reconstructive & Hand Surgery, Haga Hospital and Xpert Clinic, Den Haag, The Netherlands
| | - Carlijn R. Hooijmans
- Department for Health Evidence Unit SYRCLE, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Anesthesiology, Pain and Palliative Care (Meta Research Team), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dominic M. Power
- Department of Hand & Peripheral Nerve Surgery, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - J. Michiel Zuidam
- Department of Plastic & Reconstructive Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Tim De Jong
- Department of Plastic & Reconstructive Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Lewis M, David G, Jacobs D, Kuczwara P, Woessner AE, Kim JW, Quinn KP, Song Y. Neuro-regenerative behavior of adipose-derived stem cells in aligned collagen I hydrogels. Mater Today Bio 2023; 22:100762. [PMID: 37600354 PMCID: PMC10433000 DOI: 10.1016/j.mtbio.2023.100762] [Citation(s) in RCA: 7] [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: 05/14/2023] [Revised: 07/16/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
Peripheral nerve injuries persist as a major clinical issue facing the US population and can be caused by stretch, laceration, or crush injuries. Small nerve gaps are simple to treat, and the nerve stumps can be reattached with sutures. In longer nerve gaps, traditional treatment options consist of autografts, hollow nerve guidance conduits, and, more recently, manufactured fibrous scaffolds. These manufactured scaffolds often incorporate stem cells, growth factors, and/or extracellular matrix (ECM) proteins to better mimic the native environment but can have issues with homogenous cell distribution or uniformly oriented neurite outgrowth in scaffolds without fibrous alignment. Here, we utilize a custom device to fabricate collagen I hydrogels with aligned fibers and encapsulated adipose-derived mesenchymal stem cells (ASCs) for potential use as a peripheral nerve repair graft. Initial results of our scaffold system revealed significantly less cell viability in higher collagen gel concentrations; 3 mg/mL gels showed 84.8 ± 7.3% viable cells, compared to 6 mg/mL gels viability of 76.7 ± 9.5%. Mechanical testing of the 3 mg/mL gels showed a Young's modulus of 6.5 ± 0.8 kPa nearly matching 7.45 kPa known to support Schwann cell migration. Further analysis of scaffolds coupled with stretching in vitro revealed heightened angiogenic and factor secretion, ECM deposition, fiber alignment, and dorsal root ganglia (DRG) neurite outgrowth along the axis of fiber alignment. Our platform serves as an in vitro testbed to assess neuro-regenerative potential of ASCs in aligned collagen fiber scaffolds and may provide guidance on next-generation nerve repair scaffold design.
Collapse
Affiliation(s)
- Mackenzie Lewis
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Gabriel David
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Danielle Jacobs
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Patrick Kuczwara
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
- Department of Biological & Agricultural Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Alan E. Woessner
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Jin-Woo Kim
- Department of Biological & Agricultural Engineering; University of Arkansas, Fayetteville, AR, USA
- Materials Science & Engineering Program; University of Arkansas, Fayetteville, AR, USA
| | - Kyle P. Quinn
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Younghye Song
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
4
|
Vallejo FA, Diaz A, Errante EL, Smartz T, Khan A, Silvera R, Brooks AE, Lee YS, Burks SS, Levi AD. Systematic review of the therapeutic use of Schwann cells in the repair of peripheral nerve injuries: Advancements from animal studies to clinical trials. Front Cell Neurosci 2022; 16:929593. [PMID: 35966198 PMCID: PMC9372346 DOI: 10.3389/fncel.2022.929593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022] Open
Abstract
Objective To systematically evaluate the literature on the therapeutic use of Schwann cells (SC) in the repair of peripheral nerve injuries. Methods The Cochrane Library and PubMed databases were searched using terms [(“peripheral nerve injury” AND “Schwann cell” AND “regeneration”) OR (“peripheral nerve injuries”)]. Studies published from 2008 to 2022 were eligible for inclusion in the present study. Only studies presenting data from in-vivo investigations utilizing SCs in the repair of peripheral nerve injuries qualified for review. Studies attempting repair of a gap of ≥10 mm were included. Lastly, studies needed to have some measure of quantifiable regenerative outcome data such as histomorphometry, immunohistochemical, electrophysiology, or other functional outcomes. Results A search of the PubMed and Cochrane databases revealed 328 studies. After screening using the abstracts and methods, 17 studies were found to meet our inclusion criteria. Good SC adherence and survival in conduit tubes across various studies was observed. Improvement in morphological and functional outcomes with the use of SCs in long gap peripheral nerve injuries was observed in nearly all studies. Conclusion Based on contemporary literature, SCs have demonstrated clear potential in the repair of peripheral nerve injury in animal studies. It has yet to be determined which nerve conduit or graft will prove superior for delivery and retention of SCs for nerve regeneration. Recent developments in isolation and culturing techniques will enable further translational utilization of SCs in future clinical trials.
Collapse
Affiliation(s)
- Frederic A. Vallejo
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anthony Diaz
- Department of Neurosurgery, University of Connecticut, Farmington, CT, United States
| | - Emily L. Errante
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Taylor Smartz
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Aisha Khan
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Risset Silvera
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Adriana E. Brooks
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Yee-Shuan Lee
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Stephen Shelby Burks
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Allan D. Levi
- Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- *Correspondence: Allan D. Levi
| |
Collapse
|
5
|
Abstract
BACKGROUND Acellular nerve allograft (ANA) occupies an increasingly prominent role in the treatment of peripheral nerve reconstruction. There is demonstrable efficacy; however, some grafts fail to support axonal regrowth and the reasons for this are unclear. This study examines the ANA experience in a specialized peripheral nerve surgery department to discuss the clinical and histological findings in failed cases. METHOD Failed ANA grafts were identified from a prospective database using Medical Research Council Classification (MRCC) S3 and M3 as thresholds for success. Cases in which ANA grafting was indicated for nerve related pain and dysesthesia but where no subjective improvement in symptoms occurred were also included. Patients requiring revision surgery after ANA grafting were also considered failures. Cases were then examined in conjunction with a literature review to identify possible mechanisms of failure, including detailed histological analysis in 2 cases. RESULTS Eight failed procedures were identified from a database of 99 separate allograft records on 74 patients. This included procedures for 2 tibial nerves, 2 superficial radial nerves, 2 median nerves, 1 digital nerve and a lateral cord brachial plexus injury (male/female, 5:3; age range, 24-54 years). Allograft length range 25 to 120 mm. One postoperative infection was identified. Histological findings in 2 cases included adequate vascularization of allograft material without subsequent axonal regeneration, a reduction of large myelinated fibers proximal to a tibial nerve allograft in the setting of a chronic injury, and a preference for small rather than large fiber regeneration. CONCLUSIONS This article reports instances of ANA graft failure in a variety of contexts, for which the primary reasons for failure remain unclear. The etiology is likely to be multifactorial with both patient, graft and surgeon factors contributing to failure. Further clinical and histological analysis of ANA failures will improve our understanding of the mechanisms of graft failure.
Collapse
Affiliation(s)
- Calum Thomson
- From the Department of Peripheral Nerve Surgery, Queen Elizabeth Hospital
| | | | - Ute Pohl
- Department of Cellular Pathology
| | - Dominic M Power
- The Birmingham Peripheral Nerve Injury Service, Queen Elizabeth Hospital, Birmingham, United Kingdom
| |
Collapse
|
6
|
Im JH, Shin SH, Lee MK, Lee SR, Lee JJ, Chung YG. Evaluation of anatomical and histological characteristics of human peripheral nerves: as an effort to develop an efficient allogeneic nerve graft. Cell Tissue Bank 2022; 23:591-606. [DOI: 10.1007/s10561-022-09998-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/13/2022] [Indexed: 12/14/2022]
|
7
|
Wang Q, Chen FY, Ling ZM, Su WF, Zhao YY, Chen G, Wei ZY. The Effect of Schwann Cells/Schwann Cell-Like Cells on Cell Therapy for Peripheral Neuropathy. Front Cell Neurosci 2022; 16:836931. [PMID: 35350167 PMCID: PMC8957843 DOI: 10.3389/fncel.2022.836931] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/02/2022] [Indexed: 12/11/2022] Open
Abstract
Peripheral neuropathy is a common neurological issue that leads to sensory and motor disorders. Over time, the treatment for peripheral neuropathy has primarily focused on medications for specific symptoms and surgical techniques. Despite the different advantages of these treatments, functional recovery remains less than ideal. Schwann cells, as the primary glial cells in the peripheral nervous system, play crucial roles in physiological and pathological conditions by maintaining nerve structure and functions and secreting various signaling molecules and neurotrophic factors to support both axonal growth and myelination. In addition, stem cells, including mesenchymal stromal cells, skin precursor cells and neural stem cells, have the potential to differentiate into Schwann-like cells to perform similar functions as Schwann cells. Therefore, accumulating evidence indicates that Schwann cell transplantation plays a crucial role in the resolution of peripheral neuropathy. In this review, we summarize the literature regarding the use of Schwann cell/Schwann cell-like cell transplantation for different peripheral neuropathies and the potential role of promoting nerve repair and functional recovery. Finally, we discuss the limitations and challenges of Schwann cell/Schwann cell-like cell transplantation in future clinical applications. Together, these studies provide insights into the effect of Schwann cells/Schwann cell-like cells on cell therapy and uncover prospective therapeutic strategies for peripheral neuropathy.
Collapse
Affiliation(s)
- Qian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fang-Yu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhuo-Min Ling
- Medical School of Nantong University, Nantong, China
| | - Wen-Feng Su
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ya-Yu Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Gang Chen,
| | - Zhong-Ya Wei
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Zhong-Ya Wei,
| |
Collapse
|
8
|
Suss PH, Ribeiro VST, Motooka CE, de Melo LC, Tuon FF. Comparative study of decellularization techniques to obtain natural extracellular matrix scaffolds of human peripheral-nerve allografts. Cell Tissue Bank 2021; 23:511-520. [PMID: 34767141 DOI: 10.1007/s10561-021-09977-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND We hypothesize that adding sonication cycles to the process of decellularization of cadaveric human peripheral nerves will increase the removal of cell debris and myelin sheath, increasing their utility as allografts. METHODS Our aim of this study was to develop a decellularization protocol that allows the removal of cells and myelin sheath without detrimental effects on nerve architecture. Segments of ulnar and median nerves from human donors, isolated both before and after cardiac arrest, were subjected to two methods of decellularization: two-detergent-based (M1) and the same method with sonication added (M2). We evaluated the histology of unprocessed and decellularized nerves (n = 24 per group) for general morphology, presence of cell nuclei, nuclear remnants, collagen fibers, and myelin. We performed immunohistochemistry to verify the removal of Schwann cells associated with histomorphometry. We used scanning electron microscopy (EM) to evaluate the ultrastructure of both native and decellularized nerves. The efficacy of decellularization was assessed by analysis of genomic DNA. RESULTS Histology confirmed that both decellularization protocols were adequate and maintained natural nerve architecture. Scanning EM showed that 3D ultrastructural architecture also was maintained. Histomorphometric parameters showed a more complete removal of the myelin with the M2 protocol than with M1 (p = 0.009). Fiber diameter and density were not modified by decellularization methods. CONCLUSIONS Sonication can be a complementary method to decellularization of peripheral nerve allografts with sonication increasing the effectiveness of detergent-based protocols for the removal of unwanted cellular components from peripheral nerve allografts.
Collapse
Affiliation(s)
- Paula Hansen Suss
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Victoria Stadler Tasca Ribeiro
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Carlos Eduardo Motooka
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Letícia Corso de Melo
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil.
| |
Collapse
|
9
|
Singh S, Srivastava AK, Baranwal AK, Bhatnagar A, Das KK, Jaiswal S, Behari S. Efficacy of Silicone Conduit in the Rat Sciatic Nerve Repair Model: Journey of a Thousand Miles. Neurol India 2021; 69:318-325. [PMID: 33904443 DOI: 10.4103/0028-3886.314576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background A lot of options have been tried for bridging the two ends of the injured nerves. Researchers have used decellularized nerve grafts, artificial materials and even nerve growth factors to augment functional recovery. These materials are either costly or inaccessible in developing world. Objective The study aimed to evaluate the efficacy of the silicone conduit in a rat sciatic nerve injury model. Materials and Methods 24 healthy Sprague-Dawley (SD) rats (250-300 grams; 8-10 weeks) were used and right sciatic nerve was exposed; transected and re-anastomosed by two different methods in 16 rats. In control group, n = 8 (Group I) the sciatic nerve was untouched; Group II (reverse nerve anastomosis, n = 8): 1-centimeter of nerve was cut and re-anastomosed by using 10-0 monofilament suture; Group III (silicone conduit, n = 8) 1-centimeter nerve segment was cut, replaced by silicone conduit and supplemented by fibrin glue]. Evaluation of nerve recovery was done functionally (pain threshold and sciatic functional index) over 3 months and histologically and electron microscopically. Results Functional results showed a trend of clinical improvement in Group III and II but recovery was poor and never reached up to normal. Histopathological and electron microscopic results showed an incomplete axonal regeneration in Groups II and III. Psychological analyses showed that no outwards signs of stress were present and none of the rats showed paw biting and teeth chattering. Conclusion The silicone conduit graft may be an economical and effective alternative to presently available interposition grafts, however for short segments only.
Collapse
Affiliation(s)
- Suyash Singh
- Department of Neurosurgery, All India Institute of Medical Sciences, Raebareli, Uttar Pradesh, India
| | - Arun Kumar Srivastava
- Department of Neurosurgery, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Atul K Baranwal
- Veterinary Scientist, Animal House, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Ankur Bhatnagar
- Department of Plastic and Reconstruction Surgery, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Kuntal Kanti Das
- Department of Neurosurgery, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Sushila Jaiswal
- Department of Pathology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Sanjay Behari
- Department of Neurosurgery, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
10
|
Fuertes-Alvarez S, Izeta A. Terminal Schwann Cell Aging: Implications for Age-Associated Neuromuscular Dysfunction. Aging Dis 2021; 12:494-514. [PMID: 33815879 PMCID: PMC7990373 DOI: 10.14336/ad.2020.0708] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Action potential is transmitted to muscle fibers through specialized synaptic interfaces called neuromuscular junctions (NMJs). These structures are capped by terminal Schwann cells (tSCs), which play essential roles during formation and maintenance of the NMJ. tSCs are implicated in the correct communication between nerves and muscles, and in reinnervation upon injury. During aging, loss of muscle mass and strength (sarcopenia and dynapenia) are due, at least in part, to the progressive loss of contacts between muscle fibers and nerves. Despite the important role of tSCs in NMJ function, very little is known on their implication in the NMJ-aging process and in age-associated denervation. This review summarizes the current knowledge about the implication of tSCs in the age-associated degeneration of NMJs. We also speculate on the possible mechanisms underlying the observed phenotypes.
Collapse
Affiliation(s)
- Sandra Fuertes-Alvarez
- 1Biodonostia, Tissue Engineering Group, Paseo Dr. Begiristain, s/n, San Sebastian 20014, Spain
| | - Ander Izeta
- 1Biodonostia, Tissue Engineering Group, Paseo Dr. Begiristain, s/n, San Sebastian 20014, Spain.,2Tecnun-University of Navarra, School of Engineering, Department of Biomedical Engineering and Science, Paseo Mikeletegi, 48, San Sebastian 20009, Spain
| |
Collapse
|
11
|
Li T, Javed R, Ao Q. Xenogeneic Decellularized Extracellular Matrix-based Biomaterials For Peripheral Nerve Repair and Regeneration. Curr Neuropharmacol 2021; 19:2152-2163. [PMID: 33176651 PMCID: PMC9185777 DOI: 10.2174/1570159x18666201111103815] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/18/2021] [Accepted: 04/03/2021] [Indexed: 12/04/2022] Open
Abstract
Peripheral nerve injury could lead to either impairment or a complete loss of function for affected patients, and a variety of nerve repair materials have been developed for surgical approaches to repair it. Although autologous or autologous tissue-derived biomaterials remain preferred treatment for peripheral nerve injury, the lack of donor sources has led biomedical researchers to explore more other biomaterials. As a reliable alternative, xenogeneic decellularized extracellular matrix (dECM)-based biomaterials have been widely employed for surgical nerve repair. The dECM derived from animal donors is an attractive and unlimited source for xenotransplantation. Meanwhile, as an increasingly popular technique, decellularization could retain a variety of bioactive components in native ECM, such as polysaccharides, proteins, and growth factors. The resulting dECM-based biomaterials preserve a tissue's native microenvironment, promote Schwann cells proliferation and differentiation, and provide cues for nerve regeneration. Although the potential of dECM-based biomaterials as a therapeutic agent is rising, there are many limitations of this material restricting its use. Herein, this review discusses the decellularization techniques that have been applied to create dECM-based biomaterials, the main components of nerve ECM, and the recent progress in the utilization of xenogeneic dECM-based biomaterials through applications as a hydrogel, wrap, and guidance conduit in nerve tissue engineering. In the end, the existing bottlenecks of xenogeneic dECM-based biomaterials and developing technologies that could be eliminated to be helpful for utilization in the future have been elaborated.
Collapse
Affiliation(s)
- Ting Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Rabia Javed
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
| | - Qiang Ao
- Department of Developmental Cell Biology, China Medical University, Shenyang, China
- Institute of Regulatory Science for Med-ical Devices, Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Shi Q, Chen Y, Li M, Zhang T, Ding S, Xu Y, Hu J, Chen C, Lu H. Designing a novel vacuum aspiration system to decellularize large-size enthesis with preservation of physicochemical and biological properties. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1364. [PMID: 33313109 PMCID: PMC7723548 DOI: 10.21037/atm-20-3661] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background Functional and rapid enthesis regeneration remains a challenge after arthroscopic rotator cuff (RC) repair. Tissue-engineering a large-size biomimetic scaffold may be an adjuvant strategy to improve this clinical dilemma. Herein, we developed an optimized protocol to decellularize large-size enthesis as scaffolds for augmenting RC tear. Methods A novel vacuum aspiration system (VAS) was set up, which can provide a negative pressure to suck out cellular substances from tissue blocks without using chemical detergents. Large-size enthesis tissue specimens were harvested from canine infraspinatus tendon (IT) insertion, and then decellularized with an optimized protocol [freeze-thaw processing followed by nuclease digestion and phosphate buffer saline (PBS) rinsing in the custom-designed VAS], or a conventional protocol (freeze-thaw processing followed by nuclease digestion and PBS rinsing), thus fabricating two kinds of acellular enthesis matrix (AEM), namely C-AEM and O-AEM. After that, the C-AEM and O-AEM were comparatively evaluated from the aspect of their physicochemical and biological properties. Results Physiochemically, the O-AEM preserved the morphologies, ingredients, and tensile properties much better than the C-AEM. Biologically, in vitro studies demonstrated that both C-AEM and O-AEM show no cytotoxicity and low immunogenicity, which could promote stem cells attachment and proliferation. Interestingly, O-AEM showed better region-specific inducibility on the interacted stem cell down osteogenic, chondrogenic and tenogenic lineages compared with C-AEM. Additionally, using a canine IT repair model, the injured enthesis patched with O-AEM showed a significant improvement compared with the injured enthesis patched with C-AEM or direct suture histologically. Conclusions The proposed VAS may help us fabricate large-size AEM with good physicochemical and biological properties, and this AEM may have potential clinical applications in patching large/massive RC tear.
Collapse
Affiliation(s)
- Qiang Shi
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Yang Chen
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Muzhi Li
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Tao Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Shulin Ding
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Xu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Jianzhong Hu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Chen
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
13
|
Rhode SC, Beier JP, Ruhl T. Adipose tissue stem cells in peripheral nerve regeneration-In vitro and in vivo. J Neurosci Res 2020; 99:545-560. [PMID: 33070351 DOI: 10.1002/jnr.24738] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
After peripheral nerve injury, Schwann cells (SCs) are crucially involved in several steps of the subsequent regenerative processes, such as the Wallerian degeneration. They promote lysis and phagocytosis of myelin, secrete numbers of neurotrophic factors and cytokines, and recruit macrophages for a biological debridement. However, nerve injuries with a defect size of >1 cm do not show proper tissue regeneration and require a surgical nerve gap reconstruction. To find a sufficient alternative to the current gold standard-the autologous nerve transplant-several cell-based therapies have been developed and were experimentally investigated. One approach aims on the use of adipose tissue stem cells (ASCs). These are multipotent mesenchymal stromal cells that can differentiate into multiple phenotypes along the mesodermal lineage, such as osteoblasts, chondrocytes, and myocytes. Furthermore, ASCs also possess neurotrophic features, that is, they secrete neurotrophic factors like the nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, ciliary neurotrophic factor, glial cell-derived neurotrophic factor, and artemin. They can also differentiate into the so-called Schwann cell-like cells (SCLCs). These cells share features with naturally occurring SCs, as they also promote nerve regeneration in the periphery. This review gives a comprehensive overview of the use of ASCs in peripheral nerve regeneration and peripheral nerve tissue engineering both in vitro and in vivo. While the sustainability of differentiation of ASCs to SCLCs in vivo is still questionable, ASCs used with different nerve conduits, such as hydrogels or silk fibers, have been shown to promote nerve regeneration.
Collapse
Affiliation(s)
- Sophie Charlotte Rhode
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Justus Patrick Beier
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery and Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
14
|
Zhou LN, Wang JC, Zilundu PLM, Wang YQ, Guo WP, Zhang SX, Luo H, Zhou JH, Deng RD, Chen DF. A comparison of the use of adipose-derived and bone marrow-derived stem cells for peripheral nerve regeneration in vitro and in vivo. Stem Cell Res Ther 2020; 11:153. [PMID: 32272974 PMCID: PMC7147018 DOI: 10.1186/s13287-020-01661-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/15/2020] [Accepted: 03/23/2020] [Indexed: 01/08/2023] Open
Abstract
Background To date, it has repeatedly been demonstrated that infusing bone marrow-derived stem cells (BMSCs) into acellular nerve scaffolds can promote and support axon regeneration through a peripheral nerve defect. However, harvesting BMSCs is an invasive and painful process fraught with a low cellular yield. Methods In pursuit of alternative stem cell sources, we isolated stem cells from the inguinal subcutaneous adipose tissue of adult Sprague–Dawley rats (adipose-derived stem cells, ADSCs). We used a co-culture system that allows isolated adult mesenchymal stem cells (MSCs) and Schwann cells (SCs) to grow in the same culture medium but without direct cellular contact. We verified SC phenotype in vitro by cell marker analysis and used red fluorescent protein-tagged ADSCs to detect their fate after being injected into a chemically extracted acellular nerve allograft (CEANA). To compare the regenerative effects of CEANA containing either BMSCs or ADSCs with an autograft and CEANA only on the sciatic nerve defect in vivo, we performed histological and functional assessments up to 16 weeks after grafting. Results In vitro, we observed reciprocal beneficial effects of ADSCs and SCs in the ADSC–SC co-culture system. Moreover, ADSCs were able to survive in CEANA for 5 days after in vitro implantation. Sixteen weeks after grafting, all results consistently showed that CEANA infused with BMSCs or ADSCs enhanced injured sciatic nerve repair compared to the acellular CEANA-only treatment. Furthermore, their beneficial effects on sciatic injury regeneration were comparable as histological and functional parameters evaluated showed no statistically significant differences. However, the autograft group was roundly superior to both the BMSC- or ADSC-loaded CEANA groups. Conclusion The results of the present study show that ADSCs are a viable alternative stem cell source for treating sciatic nerve injury in lieu of BMSCs.
Collapse
Affiliation(s)
- Li Na Zhou
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China.
| | - Jia Chuan Wang
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | | | - Ya Qiong Wang
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wen Ping Guo
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Sai Xia Zhang
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Hui Luo
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Jian Hong Zhou
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Ru Dong Deng
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Dong Feng Chen
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
15
|
Pedrini FA, Boriani F, Bolognesi F, Fazio N, Marchetti C, Baldini N. Cell-Enhanced Acellular Nerve Allografts for Peripheral Nerve Reconstruction: A Systematic Review and a Meta-Analysis of the Literature. Neurosurgery 2020; 85:575-604. [PMID: 30247648 DOI: 10.1093/neuros/nyy374] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/18/2018] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Peripheral nerve reconstruction is a difficult problem to solve. Acellular nerve allografts (ANAs) have been widely tested and are a promising alternative to the autologous gold standard. However, current reconstructive methods still yield unpredictable and unsuccessful results. Consequently, numerous studies have been carried out studying alternatives to plain ANAs, but it is not clear if nerve regeneration potential exists between current biological, chemical, and physical enrichment modes. OBJECTIVE To systematically review the effects of cell-enhanced ANAs on regeneration of peripheral nerve injuries. METHODS PubMed, ScienceDirect, Medline, and Scopus databases were searched for related articles published from 2007 to 2017. Inclusion criteria of selected articles consisted of (1) articles written in English; (2) the topic being cell-enhanced ANAs in peripheral nerve regeneration; (3) an in vivo study design; and (4) postgrafting neuroregenerative assessment of results. Exclusion criteria included all articles that (1) discussed central nervous system ANAs; (2) consisted of xenografts as the main topic; and (3) consisted of case series, case reports or reviews. RESULTS Forty papers were selected, and categorization included the animal model; the enhancing cell types; the decellularization method; and the neuroregenerative test performed. The effects of using diverse cellular enhancements combined with ANAs are discussed and also compared with the other treatments such as autologous nerve graft, and plain ANAs. CONCLUSION ANAs cellular enhancement demonstrated positive effects on recovery of nerve function. Future research should include clinical translation, in order to increase the level of evidence available on peripheral nerve reconstruction.
Collapse
Affiliation(s)
- Francesca Alice Pedrini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Filippo Boriani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Department of Plastic and Hand Surgery, Koelliker Hospital, Turin, Italy
| | - Federico Bolognesi
- Maxillofacial Surgery Unit, S. Orsola-Malpighi Hospital, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Nicola Fazio
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Claudio Marchetti
- Maxillofacial Surgery Unit, S. Orsola-Malpighi Hospital, Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Nicola Baldini
- Orthopaedic Pathophysiology and Regenerative Medicine Unit, Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Pisciotta A, Bertoni L, Vallarola A, Bertani G, Mecugni D, Carnevale G. Neural crest derived stem cells from dental pulp and tooth-associated stem cells for peripheral nerve regeneration. Neural Regen Res 2020; 15:373-381. [PMID: 31571644 PMCID: PMC6921350 DOI: 10.4103/1673-5374.266043] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 05/11/2019] [Indexed: 12/15/2022] Open
Abstract
The peripheral nerve injuries, representing some of the most common types of traumatic lesions affecting the nervous system, are highly invalidating for the patients besides being a huge social burden. Although peripheral nervous system owns a higher regenerative capacity than does central nervous system, mostly depending on Schwann cells intervention in injury repair, several factors determine the extent of functional outcome after healing. Based on the injury type, different therapeutic approaches have been investigated so far. Nerve grafting and Schwann cell transplantation have represented the gold standard treatment for peripheral nerve injuries, however these approaches own limitations, such as scarce donor nerve availability and donor site morbidity. Cell based therapies might provide a suitable tool for peripheral nerve regeneration, in fact, the ability of different stem cell types to differentiate towards Schwann cells in combination with the use of different scaffolds have been widely investigated in animal models of peripheral nerve injuries in the last decade. Dental pulp is a promising cell source for regenerative medicine, because of the ease of isolation procedures, stem cell proliferation and multipotency abilities, which are due to the embryological origin from neural crest. In this article we review the literature concerning the application of tooth derived stem cell populations combined with different conduits to peripheral nerve injuries animal models, highlighting their regenerative contribution exerted through either glial differentiation and neuroprotective/neurotrophic effects on the host tissue.
Collapse
Affiliation(s)
- Alessandra Pisciotta
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Bertoni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Vallarola
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Bertani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Mecugni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Azienda USL - Institute and Health Care (IRCCS) di Reggio Emilia, Reggio Emilia, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
17
|
Rbia N, Bulstra LF, Friedrich PF, Bishop AT, Nijhuis TH, Shin AY. Gene expression and growth factor analysis in early nerve regeneration following segmental nerve defect reconstruction with a mesenchymal stromal cell-enhanced decellularized nerve allograft. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e2579. [PMID: 32095395 PMCID: PMC7015582 DOI: 10.1097/gox.0000000000002579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/21/2019] [Indexed: 12/16/2022]
Abstract
The purpose of this study was to evaluate the molecular mechanisms underlying nerve repair by a decellularized nerve allograft seeded with adipose-derived mesenchymal stromal cells (MSCs) and compare it to the unseeded allograft and autograft nerve. METHODS Undifferentiated MSCs were seeded onto decellularized nerve allografts and used to reconstruct a 10 mm gap in a rat sciatic nerve model. Gene expression profiles of genes essential for nerve regeneration and immunohistochemical staining (IHC) for PGP9.5, NGF, RECA-1, and S100 were obtained 2 weeks postoperatively. RESULTS Semi-quantitative RT-PCR analysis showed that the angiogenic molecule VEGFA was significantly increased in seeded allografts, and transcription factor SOX2 was downregulated in seeded allografts. Seeded grafts showed a significant increase in immunohistochemical markers NGF and RECA-1, when compared with unseeded allografts. CONCLUSIONS MSCs contributed to the secretion of trophic factors. A beneficial effect of the MSCs on angiogenesis was found when compared with the unseeded nerve allograft, but implanted MSCs did not show evidence of differentiation into Schwann cell-like cells.
Collapse
Affiliation(s)
- Nadia Rbia
- From the Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minn
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Liselotte F. Bulstra
- From the Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minn
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Allen T. Bishop
- From the Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minn
| | - Tim H.J. Nijhuis
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alexander Y. Shin
- From the Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minn
| |
Collapse
|
18
|
Li Z, Zhang S, Li J, Zeng H, Wang Y, Huang Y. Nerve regeneration in rat peripheral nerve allografts: Evaluation of cold-inducible RNA-binding protein in nerve storage and regeneration. J Comp Neurol 2019; 527:2885-2895. [PMID: 31116410 DOI: 10.1002/cne.24716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 01/26/2023]
Abstract
The prevalence of peripheral nerve injury has attracted increased attention. Allografting has been proposed as a potential treatment strategy for peripheral nerve injury. Moreover, cryopreservation may provide almost unlimited graft material. We investigated whether cold-inducible RNA-binding protein (CIRP) could protect peripheral nerves during cryopreservation to promote regeneration postoperation. First, CIRP was highly expressed after pretreatment at 32°C. After 4 weeks of cryopreservation, the increased live cells, low Bax/Bcl-2 ratio and high nerve growth factor and glial cell-derived neurotrophic factor levels in the 32°C group demonstrated high nerve graft viability. At 4 weeks postoperation, 32°C-Allo group demonstrated low plasma levels of interleukin-6 and interferon-gamma and a diminished cellular immune response. At 20 weeks postoperation, nerve regeneration in the 32°C-Allo group was similar to that in the fresh isograft group and superior to that in the 4°C-Allo and 15°C-Allo groups. Moreover, the compound muscle action potential and the motor nerve conduction velocity of the 32°C-Allo group were equal to those of the fresh isograft group. In conclusion, CIRP induction increased Schwann cell biological activity, inhibited cell apoptosis, reduced immune rejection, and promoted recipient nerve regeneration. Thus, CIRP could exert protective effects during nerve storage and stimulate regeneration in peripheral nerve reconstruction.
Collapse
Affiliation(s)
- Zijian Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China.,Nanchong Hospital of Traditional Chinese Medicine, Nanchong, China
| | - Song Zhang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Jinxiu Li
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China.,Nanchong Hospital of Traditional Chinese Medicine, Nanchong, China
| | - Huanhuan Zeng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Yi Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| | - Yingru Huang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Isaacs J, Feger MA, Mallu S, Patel G, Debkowska M, Yager D, Ernst B, Chilukuri S, Moser M, Kurtz C. Side-to-side supercharging nerve allograft enhances neurotrophic potential. Muscle Nerve 2019; 61:243-252. [PMID: 31724205 DOI: 10.1002/mus.26753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Critical limitations of processed acellular nerve allograft (PNA) are linked to Schwann cell function. Side-to-side bridge grafting may enhance PNA neurotrophic potential. METHODS Sprague-Dawley rats underwent tibial nerve transection and immediate repair with 20-mm PNA (n = 33) or isograft (ISO; n = 9) or 40-mm PNA (n = 33) or ISO (n = 9). Processed acellular nerve allograft groups received zero, one, or three side-to-side bridge grafts between the peroneal nerve and graft. Muscle weight, force generation, and nerve histomorphology were tested 20 weeks after repair. Selected animals underwent neuron back labeling with fluorescent dyes. RESULTS Inner axon diameters, g-ratios, and axon counts were smaller in the distal vs proximal aspect of each graft (P < .05). Schwann cell counts were greater, with a lower proportion of senescent cells for groups with bridges (P < .05). Retrograde labeling demonstrated that 6.6% to 17.7% of reinnervating neurons were from the peroneal pool. DISCUSSION Bridge grafting positively influenced muscle recovery and Schwann cell counts and senescence after long PNA nerve reconstruction.
Collapse
Affiliation(s)
- Jonathan Isaacs
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Mark A Feger
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Satya Mallu
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Gaurangkumar Patel
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Monika Debkowska
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Dorne Yager
- Divison of Plastic Surgery, Department of General Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Brady Ernst
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Sravya Chilukuri
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Matthew Moser
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Camden Kurtz
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, Virginia
| |
Collapse
|
20
|
Philips C, Cornelissen M, Carriel V. Evaluation methods as quality control in the generation of decellularized peripheral nerve allografts. J Neural Eng 2019; 15:021003. [PMID: 29244032 DOI: 10.1088/1741-2552/aaa21a] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nowadays, the high incidence of peripheral nerve injuries and the low success ratio of surgical treatments are driving research to the generation of novel alternatives to repair critical nerve defects. In this sense, tissue engineering has emerged as a possible alternative with special attention to decellularization techniques. Tissue decellularization offers the possibility to obtain a cell-free, natural extracellular matrix (ECM), characterized by an adequate 3D organization and proper molecular composition to repair different tissues or organs, including peripheral nerves. One major problem, however, is that there are no standard quality control methods to evaluate decellularized tissues. Therefore, in this review, a brief description of current strategies for peripheral nerve repair is given, followed by an overview of different decellularization methods used for peripheral nerves. Furthermore, we extensively discuss the available and currently used methods to demonstrate the success of tissue decellularization in terms of the cell removal, preservation of essential ECM molecules and maintenance or modification of biomechanical properties. Finally, orientative guidelines for the evaluation of decellularized peripheral nerve allografts are proposed.
Collapse
Affiliation(s)
- Charlot Philips
- Tissue Engineering and Biomaterials Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, De Pintelaan 185, B-9000 Ghent, Belgium
| | | | | |
Collapse
|
21
|
Isaacs J, Patel G, Mallu S, Ugwu-Oju O, Desai A, Borschel G, David D, Protzuk O, Shah S, Semus R. Effect of Reverse End-to-Side (Supercharging) Neurotization in Long Processed Acellular Nerve Allograft in a Rat Model. J Hand Surg Am 2019; 44:419.e1-419.e10. [PMID: 30172450 DOI: 10.1016/j.jhsa.2018.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 05/06/2018] [Accepted: 07/11/2018] [Indexed: 02/02/2023]
Abstract
PURPOSE Processed acellular nerve allograft (PNA) has been suggested as a convenient tool for overcoming short and medium nerve defects. Although the clinical implications are unclear, animal data suggest that PNA becomes less effective at longer lengths. Although reverse or supercharging end-to-side nerve transfer may improve the neurotrophic potential in chronically denervated nerve tissue, the application of this strategy to long acellular nerve allograft has not been previously investigated. We hypothesized that supercharging acellular nerve allograft would increase its effective length. METHODS Sprague-Dawley and Thy1-green fluorescent protein Sprague-Dawley rats underwent transection of the tibial nerve, followed by immediate repair with 20-, 40-, or 60-mm acellular nerve allografts processed identically to commercially available human acellular nerve allograft (AxoGen, Inc., Alachua, FL) or isograft. Half of the allograft group was supercharged with a reverse end-to-side transfer from the ipsilateral peroneal nerve. At 10 weeks, the reconstructed nerve in the Thy1-green fluorescent rat groups were exposed and examined under a fluorescence-enabled microscope. At 20 weeks, the remaining rats underwent motor testing and tissue harvest for morphological examination. RESULTS In comparison with a nonenhanced allograft, supercharging had a statistically significant positive impact on the reinnervated muscle normalized force generation and distal axon counts for all graft sizes. Muscles in the supercharged group were heavier than those in the allograft group for the 40-mm-length grafts and G-ratio measurements were higher in the supercharged allograft group for 60-mm-length grafts only. CONCLUSIONS This study supports that hypothesis that supercharging nerve transfer improves axon regeneration within PNA. CLINICAL RELEVANCE When an appropriate donor nerve is available, supercharging nerve transfer may improve nerve regeneration in PNA across long nerve defects.
Collapse
Affiliation(s)
- Jonathan Isaacs
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA.
| | - Gaurangkumar Patel
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Satya Mallu
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Obinna Ugwu-Oju
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Anish Desai
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Gregory Borschel
- Division of Plastic Reconstructive Surgery, Department of Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dylan David
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Omar Protzuk
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Shalin Shah
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| | - Rachel Semus
- Division of Hand Surgery, Department of Orthopaedic Surgery, Virginia Commonwealth University Medical Center, Richmond, VA
| |
Collapse
|
22
|
Im JH, Lee JY, Lee S, Lee MG, Chung YG, Kim KW. Comparison of the regeneration induced by acellular nerve allografts processed with or without chondroitinase in a rat model. Cell Tissue Bank 2019; 20:307-319. [PMID: 31030290 DOI: 10.1007/s10561-019-09770-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/19/2019] [Indexed: 12/14/2022]
Abstract
There have been various studies about the acellular nerve allograft (ANA) as the alternative of autologous nerve graft in the treatment of peripheral nerve defects. As well as the decellularization process methods of ANA, the various enhancement methods of regeneration of the grafted ANA were investigated. The chondroitin sulfate proteoglycans (CSPGs) inhibit the action of laminin which is important for nerve regeneration in the extracellular matrix of nerve. Chondroitinase ABC (ChABC) has been reported that it enhances the nerve regeneration by degradation of CSPGs. The present study compared the regeneration of ANA between the processed without ChABC group and the processed with ChABC group in a rat sciatic nerve 15 mm gap model. At 12 weeks postoperatively, there was not a significant difference in the histomorphometric analysis. In the functional analysis, there were no significant differences in maximum isometric tetanic force, wet muscle weight of tibialis anterior. The processed without ChABC group had better result in ankle contracture angle significantly. In conclusion, there were no significant differences in the regeneration of ANA between the processed without ChABC group and the processed with ChABC group.
Collapse
Affiliation(s)
- Jin-Hyung Im
- Department of Orthopedic Surgery, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Joo-Yup Lee
- Department of Orthopedic Surgery, The Catholic University of Korea College of Medicine, Seoul, Korea.
| | | | | | - Yang-Guk Chung
- Department of Orthopedic Surgery, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Ki-Won Kim
- Department of Orthopedic Surgery, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Han GH, Peng J, Liu P, Ding X, Wei S, Lu S, Wang Y. Therapeutic strategies for peripheral nerve injury: decellularized nerve conduits and Schwann cell transplantation. Neural Regen Res 2019; 14:1343-1351. [PMID: 30964052 PMCID: PMC6524503 DOI: 10.4103/1673-5374.253511] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In recent years, the use of Schwann cell transplantation to repair peripheral nerve injury has attracted much attention. Animal-based studies show that the transplantation of Schwann cells in combination with nerve scaffolds promotes the repair of injured peripheral nerves. Autologous Schwann cell transplantation in humans has been reported recently. This article reviews current methods for removing the extracellular matrix and analyzes its composition and function. The development and secretory products of Schwann cells are also reviewed. The methods for the repair of peripheral nerve injuries that use myelin and Schwann cell transplantation are assessed. This survey of the literature data shows that using a decellularized nerve conduit combined with Schwann cells represents an effective strategy for the treatment of peripheral nerve injury. This analysis provides a comprehensive basis on which to make clinical decisions for the repair of peripheral nerve injury.
Collapse
Affiliation(s)
- Gong-Hai Han
- Kunming Medical University, Kunming, Yunnan Province; Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ping Liu
- Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Xiao Ding
- Shihezi University Medical College, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Shuai Wei
- Shihezi University Medical College, Shihezi, Xinjiang Uygur Autonomous Region, China
| | - Sheng Lu
- 920th Hospital of Joint Service Support Force, Kunming, Yunnan Province, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
24
|
Qiao W, Lu L, Wu G, An X, Li D, Guo J. DPSCs seeded in acellular nerve grafts processed by Myroilysin improve nerve regeneration. J Biomater Appl 2018; 33:819-833. [PMID: 30449254 DOI: 10.1177/0885328218812136] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Since synthetic nerve conduits do not exhibit ideal regeneration characteristics, they are generally inadequate substitutes for autologous nerve grafts in the repair of long peripheral nerve defects. To resolve this problem, in this study, a nerve regeneration acellular nerve graft (ANG) with homologous dental pulp stem cells (DPSCs) was constructed. Xenogeneic ANG was processed by Myroilysin to completely remove cells and myelin sheath, while preserving extracellular matrix (ECM) microstructure of the natural nerve. The study revealed that ANG could support cell attachment and proliferation and did not stimulate a vigorous host rejection response. After inoculation of rabbit DPSCs (r-DPSCs) onto ANG, cells were observed to align along the longitudinal axis of the acellular nerve matrix (ANM) and persistently express NGF and BDNF. Undifferentiated r-DPSCs also presented glial cell characteristics and promoted nerve regeneration after transplantation in vivo. We repaired 1 cm purebred New Zealand White Rabbits sciatic nerve defects using this nerve graft construction, and nerve gap regeneration was indicated by electrophysiological and histological analysis. Therefore, we conclude that the combination of an ANG processed by Myroilysin with DPSCs providing a microenvironment that increases nerve regeneration for repairing peripheral nerve defects.
Collapse
Affiliation(s)
- Wenlan Qiao
- Department of Orthodontics, School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, PR China
- Department of Stomatology, Qilu Hospital, and Institute of Stomatology, Shandong University, Jinan, PR China
| | - Lu Lu
- Department of Orthodontics, School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, PR China
| | - Guangxue Wu
- Department of Orthodontics, School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, PR China
| | - Xianglian An
- Department of Stomatology, Qilu Hospital, and Institute of Stomatology, Shandong University, Jinan, PR China
| | - Dong Li
- Department of Cryomedicine Lab, Qilu Hospital of Shandong University, Jinan, PR China
| | - Jing Guo
- Department of Orthodontics, School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, PR China
| |
Collapse
|
25
|
Hou B, Ye Z, Ji W, Cai M, Ling C, Chen C, Guo Y. Comparison of the Effects of BMSC-derived Schwann Cells and Autologous Schwann Cells on Remyelination Using a Rat Sciatic Nerve Defect Model. Int J Biol Sci 2018; 14:1910-1922. [PMID: 30443194 PMCID: PMC6231219 DOI: 10.7150/ijbs.26765] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 09/08/2018] [Indexed: 12/13/2022] Open
Abstract
Schwann cells (SCs) are primarily responsible for the formation of myelin sheaths, yet bone marrow mesenchymal stem cell (BMSC)-derived SCs are often used to replace autologous SCs and assist with the repair of peripheral nerve myelin sheaths. In this study, the effects of the two cell types on remyelination were compared during the repair of peripheral nerves. Methods: An acellular nerve scaffold was prepared using the extraction technique. Rat BMSCs and autologous SCs were extracted. BMSCs were induced to differentiate into BMSC-derived SCs (B-dSCs) in vitro. Seed cells (BMSCs, B-dSCs, and autologous SCs) were cocultured with nerve scaffolds (Sca) in vitro. Rats with severed sciatic nerves were used as the animal model. A composite scaffold was used to bridge the broken ends. After surgery, electrophysiology, cell tracking analyses (EdU labeling), immunofluorescence staining (myelin basic protein (MBP)), toluidine blue staining, and transmission electron microscopy were conducted to compare remyelination between the various groups and to evaluate the effects of the seed cells on myelination. One week after transplantation, only a small number of B-dSCs expressed MBP, which was far less than the proportion of MBP-expressing autologous SCs (P<0.01) but was higher than the proportion of BMSCs expressing MBP (P<0.05). Four weeks after surgery, the electrophysiology results (latency time, conductive velocity and amplitude) and various quantitative indicators of remyelination (thickness, distribution, and the number of myelinated fibers) showed that the Sca+B-dSC group was inferior to the Sca+autologous SC group (P<0.05) but was superior to the Sca+BMSC group (P<0.05). Conclusions: Within 4 weeks after surgery, the use of an acellular nerve scaffold combined with B-dSCs promotes remyelination to a certain extent, but the effect is significantly less than that of the scaffold combined with autologous SCs.
Collapse
Affiliation(s)
- Bo Hou
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Zhuopeng Ye
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Wanqing Ji
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, 510623, China
| | - Meiqin Cai
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| |
Collapse
|
26
|
Musavi L, Brandacher G, Hoke A, Darrach H, Lee WPA, Kumar A, Lopez J. Muscle-derived stem cells: important players in peripheral nerve repair. Expert Opin Ther Targets 2018; 22:1009-1016. [PMID: 30347175 DOI: 10.1080/14728222.2018.1539706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Stem cell therapy for peripheral nerve repair is a rapidly evolving field in regenerative medicine. Although most studies to date have investigated stem cells originating from bone marrow or adipose, skeletal muscle has recently been recognized as an abundant and easily accessible source of stem cells. Muscle-derived stem cells (MDSCs) are a diverse population of multipotent cells with pronounced antioxidant and regenerative capacity. Areas covered: The current literature on the various roles MDSCs serve within the micro- and macro-environment of nerve injury. Furthermore, the exciting new research that is establishing MDSC-cellular therapy as an important therapeutic modality to improve peripheral nerve regeneration. Expert opinion: MDSCs are a promising therapeutic agent for the repair of peripheral nerves; MDSCs not only undergo gliogenesis and angiogenesis, but they also orchestrate larger pro-regenerative host responses. However, the isolation, transformation, and in-vivo behavior of MDSCs require further evaluation prior to clinical application.
Collapse
Affiliation(s)
- Leila Musavi
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - Gerald Brandacher
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - Ahmet Hoke
- b The Solomon H Snyder Department of Neuroscience , Johns Hopkins University , Baltimore , Maryland
| | - Halley Darrach
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - W P Andrew Lee
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| | - Anand Kumar
- c Department of Plastic & Reconstructive Surgery , Case Western Reserve University, Rainbow Babies Children's Hospital , Cleveland , OH , USA
| | - Joseph Lopez
- a Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation Laboratory , Johns Hopkins Hospital , Baltimore , Maryland
| |
Collapse
|
27
|
Wang ZZ, Sakiyama-Elbert SE. Matrices, scaffolds & carriers for cell delivery in nerve regeneration. Exp Neurol 2018; 319:112837. [PMID: 30291854 DOI: 10.1016/j.expneurol.2018.09.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/13/2018] [Accepted: 09/28/2018] [Indexed: 12/22/2022]
Abstract
Nerve injuries can be life-long debilitating traumas that severely impact patients' quality of life. While many acellular neural scaffolds have been developed to aid the process of nerve regeneration, complete functional recovery is still very difficult to achieve, especially for long-gap peripheral nerve injury and most cases of spinal cord injury. Cell-based therapies have shown many promising results for improving nerve regeneration. With recent advances in neural tissue engineering, the integration of biomaterial scaffolds and cell transplantation are emerging as a more promising approach to enhance nerve regeneration. This review provides an overview of important considerations for designing cell-carrier biomaterial scaffolds. It also discusses current biomaterials used for scaffolds that provide permissive and instructive microenvironments for improved cell transplantation.
Collapse
Affiliation(s)
- Ze Zhong Wang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA; Department of Biomedical Engineering, University of Austin at Texas, Austin, TX, USA
| | | |
Collapse
|
28
|
Hou B, Cai M, Chen C, Ji W, Ye Z, Ling C, Chen Z, Guo Y. Xenogeneic acellular nerve scaffolds supplemented with autologous bone marrow-derived stem cells promote axonal outgrowth and remyelination but not nerve function. J Biomed Mater Res A 2018; 106:3065-3078. [PMID: 30260554 DOI: 10.1002/jbm.a.36497] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 06/07/2018] [Accepted: 06/27/2018] [Indexed: 12/29/2022]
Abstract
Autologous nerves, artificial scaffolds or acellular nerve scaffolds are commonly used in bridging treatment for peripheral nerve defects. Xenogeneic acellular nerve scaffolds and allogeneic cellular nerve scaffolds have the same structural characteristics. Due to the wider source of raw materials, these latter scaffolds have high-potential value for applications. However, whether their heterogeneity will affect nerve regeneration is unknown. The current study evaluated the efficiency of xenogeneic acellular nerve scaffolds (XANs) combined with 5-ethynyl-2'-deoxyuridine (EdU)-labeling of autologous bone marrow-derived stem cells (BMSCs) for repair of a 1.5 cm gap in rat sciatic nerves. XANs from rabbit tibial nerves were prepared, the structure and components of the scaffolds were evaluated after completely removing the cellular components. Animals were divided into four groups based on graft: the simple XAN group, the XAN + BMSC group, the XAN + Media (from BMSC culture) group, and the autograft group. Serological immune tests showed that XANs induce an immune response in the first 2 weeks after transplantation. Moreover, cell tracking revealed that the proportion of EdU+ cells decreased over time, as shown by the measures at 2 days (70%), 4 days (20%), and 8 days (even <3%) postoperatively. Nerve functional analyses revealed that in contrast to the autograft group results, the XAN-BMSC, XAN + Media, and XAN groups did not exhibit good restoration of the sciatic functional index (SFI) or electrophysiological results (the peak action potential amplitudes) 12 weeks, postoperatively. However, the XAN-BMSC and autograft groups demonstrated greater remyelination and increased axon numbers and myelin thickness than the XAN + Media and XAN groups 12 weeks, postoperatively (p < .05). In conclusion, in the early stage of transplantation, XANs induce a certain degree of inflammation. Although the combination of XANs with autologous BMSCs enhanced the number of regenerated axons and the remyelination, the combination did not effectively improve the recovery of nervous motor function. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3065-3078, 2018.
Collapse
Affiliation(s)
- Bo Hou
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| | - Meiqin Cai
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| | - Chuan Chen
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| | - Wanqing Ji
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Province, 510623, Guangzhou, China
| | - Zhuopeng Ye
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| | - Cong Ling
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| | - Zhuopeng Chen
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| | - Ying Guo
- Departments of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangdong Province, 510630, Guangzhou, China
| |
Collapse
|
29
|
Salehi M, Naseri-Nosar M, Ebrahimi-Barough S, Nourani M, Khojasteh A, Farzamfar S, Mansouri K, Ai J. Polyurethane/Gelatin Nanofibrils Neural Guidance Conduit Containing Platelet-Rich Plasma and Melatonin for Transplantation of Schwann Cells. Cell Mol Neurobiol 2018; 38:703-713. [PMID: 28823058 DOI: 10.1007/s10571-017-0535-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/08/2017] [Indexed: 10/19/2022]
Abstract
The current study aimed to enhance the efficacy of peripheral nerve regeneration using a biodegradable porous neural guidance conduit as a carrier to transplant allogeneic Schwann cells (SCs). The conduit was prepared from polyurethane (PU) and gelatin nanofibrils (GNFs) using thermally induced phase separation technique and filled with melatonin (MLT) and platelet-rich plasma (PRP). The prepared conduit had the porosity of 87.17 ± 1.89%, the contact angle of 78.17 ± 5.30° and the ultimate tensile strength and Young's modulus of 5.40 ± 0.98 MPa and 3.13 ± 0.65 GPa, respectively. The conduit lost about 14% of its weight after 60 days in distilled water. The produced conduit enhanced the proliferation of SCs demonstrated by a tetrazolium salt-based assay. For functional analysis, the conduit was seeded with 1.50 × 104 SCs (PU/GNFs/PRP/MLT/SCs) and implanted into a 10-mm sciatic nerve defect of Wistar rat. Three control groups were used: (1) PU/GNFs/SCs, (2) PU/GNFs/PRP/SCs, and (3) Autograft. The results of sciatic functional index, hot plate latency, compound muscle action potential amplitude and latency, weight-loss percentage of wet gastrocnemius muscle and histopathological examination using hematoxylin-eosin and Luxol fast blue staining, demonstrated that using the PU/GNFs/PRP/MLT conduit to transplant SCs to the sciatic nerve defect resulted in a higher regenerative outcome than the PU/GNFs and PU/GNFs/PRP conduits.
Collapse
Affiliation(s)
- Majid Salehi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P.O. Box 1417755469, Tehran, Iran
| | - Mahdi Naseri-Nosar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P.O. Box 1417755469, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P.O. Box 1417755469, Tehran, Iran
| | - Mohammdreza Nourani
- Nano Biotechnology Research Center, Baqiyatallah University of Medical Sciences, P.O. Box 1435944711, Tehran, Iran
| | - Arash Khojasteh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, P.O. Box 1983969411, Tehran, Iran
| | - Saeed Farzamfar
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P.O. Box 1417755469, Tehran, Iran
| | - Korosh Mansouri
- Department of Physical Medicine and Rehabilitation, Iran University of Medical Sciences, P.O. Box 14665354, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P.O. Box 1417755469, Tehran, Iran.
| |
Collapse
|
30
|
Mackenzie SJ, Yi JL, Singla A, Russell TM, Osterhout DJ, Calancie B. Cauda equina repair in the rat: Part 3. Axonal regeneration across Schwann cell-Seeded collagen foam. Muscle Nerve 2017; 57:E78-E84. [PMID: 28746726 DOI: 10.1002/mus.25751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/12/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Treatments for patients with cauda equina injury are limited. METHODS In this study, we first used retrograde labeling to determine the relative contributions of cauda equina motor neurons to intrinsic and extrinsic rat tail muscles. Next, we transected cauda equina ventral roots and proceeded to bridge the proximal and distal stumps with either a type I collagen scaffold coated in laminin (CL) or a collagen-laminin scaffold that was also seeded with Schwann cells (CLSC). Regeneration was assessed by way of serial retrograde labeling. RESULTS After accounting for the axonal contributions to intrinsic vs. extrinsic tail muscles, we noted a higher degree of double labeling in the CLSC group (58.0 ± 39.6%) as compared with the CL group (27.8 ± 16.0%; P = 0.02), but not the control group (33.5 ± 18.2%; P = 0.10). DISCUSSION Our findings demonstrate the feasibility of using CLSCs in cauda equina injury repair. Muscle Nerve 57: E78-E84, 2018.
Collapse
Affiliation(s)
- Samuel J Mackenzie
- Department of Neuroscience, Upstate Medical University, Syracuse, New York, USA
| | - Juneyoung L Yi
- Department of Neurosurgery, Upstate Medical University, IHP 1213, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Amit Singla
- Department of Neurosurgery, Upstate Medical University, IHP 1213, 750 East Adams Street, Syracuse, New York, 13210, USA
| | - Thomas M Russell
- Department of Cell and Developmental Biology, Upstate Medical University, Syracuse, New York, USA
| | - Donna J Osterhout
- Department of Cell and Developmental Biology, Upstate Medical University, Syracuse, New York, USA
| | - Blair Calancie
- Department of Neurosurgery, Upstate Medical University, IHP 1213, 750 East Adams Street, Syracuse, New York, 13210, USA
| |
Collapse
|
31
|
Micropuncture and pressure assisted Schwann cell seeding of nerve allograft. J Neurosci Methods 2017; 287:47-52. [PMID: 28606481 DOI: 10.1016/j.jneumeth.2017.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Tissue processing to create immunotolerant nerve allograft removes neurosupportive cells. Few strategies have been described for implanting new cells into the graft to support axonal regeneration. NEW METHOD Micropuncture of the nerve allograft surface combined with immersion into a pressurized cell-rich solution to potentiate the introduction of viable Schwann cells (SC) into processed nerve allograft. Allografts were used to repair rodent sciatic nerve defects. At 3, 7, and 21days, grafts were harvested, stained for SCs, and analyzed using total cross sectional area (CSA) occupied by SCs to quantify SC presence. RESULTS At days 3 and 7, SC CSA was significantly greater for the injection group compared to all other groups. At day 21, SC CSA for the injection group (0.2252%±0.2730) was significantly greater compared to following groups: pressurized-punctured (0.0653%±0.0934), nonpressurized-nonpunctured (0.0607%±0.0709), punctured-control (0.0246%±0.0398), and nonpunctured-control (0.0126%±0.0151). A significant decrease in percent CSA occupied by SCs from day 3 to day 21 was noted in nonpressurized-punctured group (p=0.0106), pressurized-nonpunctured group (p=0.0477), and injection group (p=0.0010). COMPARISON WITH EXISTING METHOD(S) Most studies have used small caliber hypodermic needles to inject the cells into grafts. CONCLUSIONS Despite a presumed decrease in cell viability over the three weeks of the study, the large initial inoculum achieved by injection technique results in higher levels of final SC seeding in acellular nerve allograft compared with bathing techniques with or without micropuncture or pressurization.
Collapse
|
32
|
Xu K, Kuntz LA, Foehr P, Kuempel K, Wagner A, Tuebel J, Deimling CV, Burgkart RH. Efficient decellularization for tissue engineering of the tendon-bone interface with preservation of biomechanics. PLoS One 2017; 12:e0171577. [PMID: 28170430 PMCID: PMC5295703 DOI: 10.1371/journal.pone.0171577] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/22/2017] [Indexed: 02/07/2023] Open
Abstract
Interfaces between tendon/ligament and bone (“entheses”) are highly specialized tissues that allow for stress transfer between mechanically dissimilar materials. Entheses show very low regenerative capacity resulting in high incidences of failure after surgical repair. Tissue engineering is a promising approach to recover functionality of entheses. Here, we established a protocol to decellularize porcine entheses as scaffolds for enthesis tissue engineering. Chemical detergents as well as physical treatments were investigated with regard to their efficiency to decellularize 2 mm thick porcine Achilles tendon entheses. A two-phase approach was employed: study 1 investigated the effect of various concentrations of sodium dodecyl sulfate (SDS) and t-octylphenoxypolyethoxy-ethanol (Triton X-100) as decellularization agents. The most efficient combination of SDS and Triton was then carried forward into study 2, where different physical methods, including freeze-thaw cycles, ultrasound, perfusion, and hydrostatic washing were used to enhance the decellularization effect. Cell counts, DNA quantification, and histology showed that washing with 0.5% SDS + 1% Triton X-100 for 72 h at room temperature could remove ~ 98% cells from the interface. Further investigation of physical methods proved that washing under 200 mmHg hydrostatic pressure shortened the detergent exposing time from 72 h to 48 h. Biomechanical tensile testing showed that the biomechanical features of treated samples were preserved. Washing under 200 mmHg hydrostatic pressure with 0.5% SDS + 1% Triton X-100 for 48 h efficiently decellularized entheses with preservation of matrix structure and biomechanical features. This protocol can be used to efficiently decellularize entheses as scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Kai Xu
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany.,Department of Orthopedics, Tongji Hospital, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Lara A Kuntz
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Peter Foehr
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Katharina Kuempel
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Alexandra Wagner
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Jutta Tuebel
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Constantin V Deimling
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Rainer H Burgkart
- Department of Orthopaedics and Sportsorthopaedics, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
33
|
Thompson MJ, Patel G, Isaacs J, McMurtry J, Richards N, Daner W. Introduction of neurosupportive cells into processed acellular nerve allografts results in greater number and more even distribution when injected compared to soaking techniques. Neurol Res 2017; 39:189-197. [PMID: 28112028 DOI: 10.1080/01616412.2017.1282336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES Processing necessary to remove immunogenic components of nerve allograft renders it acellular. Seeding with supportive cells may improve axon regeneration. We aim to identify the method associated with implantation of the greatest volume and most even distribution of cells. METHODS Hypodermic needle injection was compared to soaking in solution under both normal and pressurized conditions after micropuncture of the allograft. Distribution within the allograft was measured using an in vitro model of fluorescent beads, as well as cultured Schwann cells. RESULTS Injection treatment resulted in larger volumes and a more uniform cross-sectional distribution of implanted cells. Beads and cells behaved similarly relative to the measured outcomes. CONCLUSIONS Injection instills more cells in a more uniform distribution. In vivo testing may evaluate whether these techniques vary relative to cell survival, cell migration, and clinical outcomes. Size- and concentration-matched fluorescent beads may represent a viable model for analyzing cell implantation.
Collapse
Affiliation(s)
- Matthew J Thompson
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - Gaurangkumar Patel
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - Jonathan Isaacs
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - John McMurtry
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - Nathan Richards
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| | - William Daner
- a Division of Hand Surgery, Department of Orthopaedic Surgery , Virginia Commonwealth University Medical Center , Richmond , VA , USA
| |
Collapse
|
34
|
Chen SL, Chen ZG, Dai HL, Ding JX, Guo JS, Han N, Jiang BG, # HJ, Li J, Li SP, Li WJ, Liu J, Liu Y, Ma JX, Peng J, Shen YD, Sun GW, Tang PF, Wang GH, Wang XH, Xiang LB, Xie RG, Xu JG, Yu B, Zhang LC, Zhang PX, Zhou SL. Repair, protection and regeneration of peripheral nerve injury. Neural Regen Res 2015; 10:1777-98. [PMID: 26807113 PMCID: PMC4705790 DOI: 10.4103/1673-5374.170301] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
35
|
Jiang LF, Chen O, Chu TG, Ding J, Yu Q. T Lymphocyte Subsets and Cytokines in Rats Transplanted with Adipose-Derived Mesenchymal Stem Cells and Acellular Nerve for Repairing the Nerve Defects. J Korean Neurosurg Soc 2015; 58:101-6. [PMID: 26361524 PMCID: PMC4564740 DOI: 10.3340/jkns.2015.58.2.101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/13/2015] [Accepted: 03/23/2015] [Indexed: 11/27/2022] Open
Abstract
Objective The aim of this study was to explore the immunity in rats transplanted with adipose-derived mesenchymal stem cells (ADSCs) and acellular nerve (ACN) for repairing sciatic nerve defects. Methods ADSCs were isolated from the adipose tissues of Wistar rats. Sprague-Dawley rats were used to establish a sciatic nerve defect model and then divided into four groups, according to the following methods : Group A, allogenic nerve graft; Group B, allograft with ACN; Group C, allograft ADSCs+ACN, and Group D, nerve autograft. Results At the day before transplantation and 3, 7, 14, and 28 days after transplantation, orbital venous blood of the Sprague-Dawley rats in each group was collected to detect the proportion of CD3+, CD4+, and CD8+ subsets using flow cytometry and to determine the serum concentration of interleukin-2 (IL-2), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) using enzyme-linked immunosorbent assay (ELISA). At each postoperative time point, the proportion of CD3+, CD4+, and CD8+ subsets and the serum concentration of IL-2, TNF-α, and IFN-γ in group C were all near to those in group B and group D, in which no statistically significant difference was observed. As compared with group A, the proportion of CD3+, CD4+, and CD8+ subsets and the serum concentration of IL-2, TNF-α, and IFN-γ were significantly reduced in group C (p<0.05). Conclusion The artificial nerve established with ADSCs and ACN has no obvious allograft rejection for repairing rat nerve defects.
Collapse
Affiliation(s)
- Liang-Fu Jiang
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Ou Chen
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang, China
| | - Ting-Gang Chu
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jian Ding
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qing Yu
- Department of Hand & Plastic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
36
|
Jiang L, Zheng Y, Chen O, Chu T, Ding J, Yu Q. Nerve defect repair by differentiated adipose-derived stem cells and chondroitinase ABC-treated acellular nerves. Int J Neurosci 2015; 126:568-576. [PMID: 26000928 DOI: 10.3109/00207454.2015.1048547] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To evaluate the effects of differentiated adipose-derived stem cells (dADSC) and chondroitinase ABC (ChABC)-treated acellular nerves (ACN) in building artificial nerves and repairing nerve defects. METHODS ADSC were isolated from the adipose tissue of Wistar rats, induced to differentiate into Schwann-like cells, and implanted into ChABC-treated ACN to repair a 15-mm sciatic nerve defect in Sprague-Dawley rats (the experimental group, group D). The control groups were an autologous nerve transplantation group (group E); ACN (group A), ChABC-treated ACN graft group (group B), and dADSC + ACN (group C). Twelve weeks after surgery, electromyography recordings, tricep surae muscle wet weight recovery rate, and axon counts were measured to evaluate the repair of peripheral nerve defects. RESULTS The nerve conduction velocity, compound muscle action potentials, tricep surae muscle wet weight recovery rate, and myelinated axon counts in the ChABC-ACN/dADSC group were significantly higher than in the other groups (P < 0.05), which were all lower than the autologous group (P < 0.05). CONCLUSIONS The combination of ChABC-treated ACN and dADSC exhibited a synergistic effect in promoting nerve regeneration, and could be an alternative for effective tissue-engineered nerves.
Collapse
Affiliation(s)
- Liangfu Jiang
- a 1Department of Hand & Plastic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Zheng
- b 2Department of Children Health Care, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ou Chen
- c 3Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Tinggang Chu
- a 1Department of Hand & Plastic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Ding
- a 1Department of Hand & Plastic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Yu
- a 1Department of Hand & Plastic Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
37
|
Zhou LN, Zhang JW, Liu XL, Zhou LH. Co-Graft of Bone Marrow Stromal Cells and Schwann Cells Into Acellular Nerve Scaffold for Sciatic Nerve Regeneration in Rats. J Oral Maxillofac Surg 2015; 73:1651-60. [DOI: 10.1016/j.joms.2015.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/06/2015] [Accepted: 02/06/2015] [Indexed: 01/21/2023]
|
38
|
Yu H, Xiang L, Xu W, Zhao B, Wang Y, Peng J, Lu S. Chondroitinase ABC improves recovery of long sciatic nerve defects. Neural Regen Res 2015; 7:61-5. [PMID: 25806060 PMCID: PMC4354120 DOI: 10.3969/j.issn.1673-5374.2012.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/24/2011] [Indexed: 01/30/2023] Open
Abstract
Sciatic nerves from allogeneic Sprague-Dawley rats were pretreated with chondroitinase ABC and were used to bridge damaged sciatic nerves in Wistar rats. Chondroitin sulfate proteoglycans were removed from the chemically extracted acellular nerves. At 3 months after grafting, the footplate pinch test result was positive in the Wistar rats. Autotomy scores decreased, and increased muscular contraction tension appeared when triceps surae muscles were stimulated. In addition, the recovery rate of wet triceps surae muscle weight increased, and the distal segment of the chondroitinase ABC-treated graft exhibited Schwann cells next to the nerve fibers. These results suggested that chondroitinase ABC pretreatment enhanced repair of long nerve defects via acellular nerve grafting.
Collapse
Affiliation(s)
- Hailong Yu
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang 110016, Liaoning Province, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang 110016, Liaoning Province, China
| | - Wenjing Xu
- Institute of Orthopedics, General Hospital of Chinese PLA, Beijing 100853, China
| | - Bin Zhao
- Institute of Orthopedics, General Hospital of Chinese PLA, Beijing 100853, China
| | - Yu Wang
- Institute of Orthopedics, General Hospital of Chinese PLA, Beijing 100853, China
| | - Jiang Peng
- Institute of Orthopedics, General Hospital of Chinese PLA, Beijing 100853, China
| | - Shibi Lu
- Institute of Orthopedics, General Hospital of Chinese PLA, Beijing 100853, China
| |
Collapse
|
39
|
Yang MM, Huang W, Jiang DM. Tetramethylpyrazine protects Schwann cells from ischemia-like injury and increases cell survival in cold ischemic rat nerves. BRAZ J PHARM SCI 2015. [DOI: 10.1590/s1984-82502015000100014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tetramethylpyrazine (TMP), a major active ingredient of Ligusticum wallichi Franchat extract (a Chinese herb), exhibits neuroprotective properties in ischemia. In this study, we assessed its protective effects on Schwann cells (SCs) by culturing them in the presence of oxygen glucose deprivation (OGD) conditions and measuring cell survival in cold ischemic rat nerves. In the OGD-induced ischemic injury model of SCs, we demonstrated that TMP treatment not only reduced OGD-induced cell viability losses, cell death, and apoptosis of SCs in a dose-dependent manner, and inhibited LDH release, but also suppressed OGD-induced downregulation of Bcl-2 and upregulation of Bax and caspase-3, as well as inhibited the consequent activation of caspase-3. In the cold ischemic nerve model, we found that prolonged cold ischemic exposure for four weeks was markedly associated with the absence of SCs, a decrease in cell viability, and apoptosis in preserved nerve segments incubated in University of Wisconsin solution (UWS) alone. However, TMP attenuated nerve segment damage by preserving SCs and antagonizing the decrease in nerve fiber viability and increase in TUNEL-positive cells in a dose-dependent manner. Collectively, our results indicate that TMP not only provides protective effects in an ischemia-like injury model of cultured rat SCs by regulating Bcl-2, Bax, and caspase-3, but also increases cell survival and suppresses apoptosis in the cold ischemic nerve model after prolonged ischemic exposure for four weeks. Therefore, TMP may be a novel and effective therapeutic strategy for preventing peripheral nervous system ischemic diseases and improving peripheral nerve storage.
Collapse
Affiliation(s)
- Ming-Ming Yang
- Chongqing Medical University, People's Republic of China
| | - Wei Huang
- Chongqing Medical University, People's Republic of China
| | | |
Collapse
|
40
|
Kim DY, Choi YS, Kim SE, Lee JH, Kim SM, Kim YJ, Rhie JW, Jun YJ. In vivo effects of adipose-derived stem cells in inducing neuronal regeneration in Sprague-Dawley rats undergoing nerve defect bridged with polycaprolactone nanotubes. J Korean Med Sci 2014; 29 Suppl 3:S183-92. [PMID: 25473208 PMCID: PMC4248004 DOI: 10.3346/jkms.2014.29.s3.s183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/12/2014] [Indexed: 11/25/2022] Open
Abstract
There have been many attempts for regeneration of peripheral nerve injury. In this study, we examined the in vivo effects of non-differentiated and neuronal differentiated adipose-derived stem cells (ADSCs) in inducing the neuronal regeneration in the Sprague-Dawley (SD) rats undergoing nerve defect bridged with the PCL nanotubes. Then, we performed immunohistochemical and histopathologic examinations, as well as the electromyography, in three groups: the control group (14 sciatic nerves transplanted with the PCL nanotube scaffold), the experimental group I (14 sciatic nerves with the non-differentiated ADSCs at a density of 7×10(5) cells/0.1 mL) and the experimental group II (14 sciatic nerves with the neuronal differentiated ADSCs at 7×10(5) cells/0.1 mL). Six weeks postoperatively, the degree of the neuronal induction and that of immunoreactivity to nestin, MAP-2 and GFAP was significantly higher in the experimental group I and II as compared with the control group. In addition, the nerve conduction velocity (NCV) was significantly higher in the experimental group I and II as compared with the control group (P=0.021 and P=0.020, respectively). On the other hand, there was no significant difference in the NCV between the two experimental groups (P>0.05). Thus, our results will contribute to treating patients with peripheral nerve defects using PCL nanotubes with ADSCs.
Collapse
Affiliation(s)
- Dong-Yeon Kim
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| | - Yong-Seong Choi
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Kim
- Department of Orthopedic Surgery and Rare Diseases Institute, Korea University, Seoul, Korea
| | - Jung-Ho Lee
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| | - Sue-Min Kim
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| | - Young-Jin Kim
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| | - Jong-Won Rhie
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| | - Young-Joon Jun
- Department of Plastic and Reconstructive Surgery, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
41
|
Zhao F, He W, Zhang Y, Tian D, Zhao H, Yu K, Bai J. Electric stimulation and decimeter wave therapy improve the recovery of injured sciatic nerves. Neural Regen Res 2014; 8:1974-84. [PMID: 25206506 PMCID: PMC4145900 DOI: 10.3969/j.issn.1673-5374.2013.21.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/24/2013] [Indexed: 01/08/2023] Open
Abstract
Drug treatment, electric stimulation and decimeter wave therapy have been shown to promote the repair and regeneration of the peripheral nerves at the injured site. This study prepared a Mackinnon's model of rat sciatic nerve compression. Electric stimulation was given immediately after neurolysis, and decimeter wave radiation was performed at 1 and 12 weeks post-operation. Histological observation revealed that intraoperative electric stimulation and decimeter wave therapy could improve the local blood circulation of repaired sites, alleviate hypoxia of compressed nerves, and lessen adhesion of compressed nerves, thereby decreasing the formation of new entrapments and enhancing compressed nerve regeneration through an improved microenvironment for regeneration. Immunohistochemical staining results revealed that intraoperative electric stimulation and decimeter wave could promote the expression of S-100 protein. Motor nerve conduction velocity and amplitude, the number and diameter of myelinated nerve fibers, and sciatic functional index were significantly increased in the treated rats. These results verified that intraoperative electric stimulation and decimeter wave therapy contributed to the regeneration and the recovery of the functions in the compressed nerves.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Orthopedics, the First Hospital of Hebei Medical University, Shijiazhuang 050031, Hebei Province, China
| | - Wei He
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Yingze Zhang
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Dehu Tian
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Hongfang Zhao
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Kunlun Yu
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| | - Jiangbo Bai
- Department of Hand Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang 050051, Hebei Province, China
| |
Collapse
|
42
|
Abstract
Nerve conduits and acellular nerve allograft offer efficient and convenient tools for overcoming unexpected gaps during nerve repair. Both techniques offer guidance for migrating Schwann cells and axonal regeneration though utilizing very different scaffolds. The substantially greater amount of animal and clinical data published on nerve conduits is marked by wide discrepancies in results that may be partly explained by a still poorly defined critical repair gap and diameter size. The available information on acellular allografts appears more consistently positive though this tool is also hampered by a longer but also limited critical length. This article reviews the current relative literature and examines pertinent parameters for application of both acellular allograft and nerve conduits in overcoming short nerve gaps.
Collapse
|
43
|
Beigi MH, Ghasemi-Mobarakeh L, Prabhakaran MP, Karbalaie K, Azadeh H, Ramakrishna S, Baharvand H, Nasr-Esfahani MH. In vivo integration of poly(ε-caprolactone)/gelatin nanofibrous nerve guide seeded with teeth derived stem cells for peripheral nerve regeneration. J Biomed Mater Res A 2014; 102:4554-67. [PMID: 24677613 DOI: 10.1002/jbm.a.35119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/29/2014] [Accepted: 02/04/2014] [Indexed: 12/25/2022]
Abstract
Artificial nanofiber nerve guides have gained huge interest in bridging nerve gaps and associated peripheral nerve regeneration due to its high surface area, flexibility and porous structure. In this study, electrospun poly (ε-caprolactone)/gelatin (PCL/Gel) nanofibrous mats were fabricated, rolled around a copper wire and fixed by medical grade adhesive to obtain a tubular shaped bio-graft, to bridge 10 mm sciatic nerve gap in in vivo rat models. Stem cells from human exfoliated deciduous tooth (SHED) were transplanted to the site of nerve injury through the nanofibrous nerve guides. In vivo experiments were performed in animal models after creating a sciatic nerve gap, such that the nerve gap was grafted using (i) nanofiber nerve guide (ii) nanofiber nerve guide seeded with SHED (iii) suturing, while an untreated nerve gap remained as the negative control. In vitro cell culture study was carried out for primary investigation of SHED-nanofiber interaction and its viability within the nerve guides after 2 and 16 weeks of implantation time. Walking track analysis, plantar test, electrophysiology and immunohistochemistry were performed to evaluate functional recovery during nerve regeneration. Vascularization was also investigated by hematoxilin/eosine (H&E) staining. Overall results showed that the SHED seeded on nanofibrous nerve guide could survive and promote axonal regeneration in rat sciatic nerves, whereby the biocompatible PCL/Gel nerve guide with cells can support axonal regeneration and could be a promising tissue engineered graft for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Mohammad-Hossein Beigi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Materials Engineering Department, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Comparison of decellularization protocols for preparing a decellularized porcine annulus fibrosus scaffold. PLoS One 2014; 9:e86723. [PMID: 24475172 PMCID: PMC3901704 DOI: 10.1371/journal.pone.0086723] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 12/14/2013] [Indexed: 12/02/2022] Open
Abstract
Tissue-specific extracellular matrix plays an important role in promoting tissue regeneration and repair. We hypothesized that decellularized annular fibrosus matrix may be an appropriate scaffold for annular fibrosus tissue engineering. We aimed to determine the optimal decellularization method suitable for annular fibrosus. Annular fibrosus tissue was treated with 3 different protocols with Triton X-100, sodium dodecyl sulfate (SDS) and trypsin. After the decellularization process, we examined cell removal and preservation of the matrix components, microstructure and mechanical function with the treatments to determine which method is more efficient. All 3 protocols achieved decellularization; however, SDS or trypsin disturbed the structure of the annular fibrosus. All protocols maintained collagen content, but glycosaminoglycan content was lost to different degrees, with the highest content with TritonX-100 treatment. Furthermore, SDS decreased the tensile mechanical property of annular fibrosus as compared with the other 2 protocols. MTT assay revealed that the decellularized annular fibrosus was not cytotoxic. Annular fibrosus cells seeded into the scaffold showed good viability. The Triton X-100–treated annular fibrosus retained major extracellular matrix components after thorough cell removal and preserved the concentric lamellar structure and tensile mechanical properties. As well, it possessed favorable biocompatibility, so it may be a suitable candidate as a scaffold for annular fibrosus tissue engineering.
Collapse
|
45
|
Gao X, Wang Y, Chen J, Peng J. The role of peripheral nerve ECM components in the tissue engineering nerve construction. Rev Neurosci 2013; 24:443-53. [PMID: 23907421 DOI: 10.1515/revneuro-2013-0022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 07/05/2013] [Indexed: 11/15/2022]
Abstract
The extracellular matrix (ECM) is the naturally occurring substrate that provides a support structure and an attachment site for cells. It also produces a biological signal, which plays an important role in and has significant impact on cell adhesion, migration, proliferation, differentiation, and gene expression. Peripheral nerve repair is a complicated process involving Schwann cell proliferation and migration, 'bands of Büngner' formation, and newborn nerve extension. In the ECM of peripheral nerves, macromolecules are deposited among cells; these constitute the microenvironment of Schwann cell growth. Such macromolecules include collagen (I, III, IV, V), laminin, fibronectin, chondroitin sulfate proteoglycans (CSPGs), and other nerve factors. Collagen, the main component of ECM, provides structural support and guides newborn neurofilament extension. Laminin, fibronectin, CSPGs, and neurotrophic factors, are promoters or inhibitors, playing different roles in nerve repair after injury. By a chemical decellularization process, acellular nerve allografting eliminates the antigens responsible for allograft rejection and maintains most of the ECM components, which can effectively guide and enhance nerve regeneration. Thus, the composition and features of peripheral nerve ECM suggest its superiority as nerve repair material. This review focuses on the structure, function, and application in the tissue engineering nerve construction of the peripheral nerve ECM components.
Collapse
|
46
|
Zhu G, Lou W. Regeneration of facial nerve defects with xenogeneic acellular nerve grafts in a rat model. Head Neck 2013; 36:481-6. [PMID: 23729307 DOI: 10.1002/hed.23321] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Because of ease of harvest and low immunogenicity, xenogeneic acellular nerve graft (XANG) may be an alternative to autologous nerve to repair facial nerve defects. METHODS Facial nerve defects of Wistar rats were repaired by XANG, and nerve gap regeneration was investigated by electrophysiological test, horseradish peroxidase (HRP) retrograde tracing and histomorphometric analysis, as compared to autograft. RESULTS Twenty weeks after the grafting, electrophysiology showed that whisker pad muscles responded to the electrical stimuli given at the site proximal to the transplantation in 2 groups. Some HRP-labeled facial motorneurons were located on the facial nucleus of the operated side, and an abundance of myelinated axons were found at the middle of the grafts and obvious motor endplates in the target muscles in 2 groups, although they were inferior to the contralateral side in numbers. CONCLUSION XANG represents an alternative approach for the reconstruction of peripheral facial nerve defects.
Collapse
Affiliation(s)
- Guochen Zhu
- Department of Otolaryngology, Wuxi Second People's Hospital, Affiliated with Nanjing Medical University, Wuxi, Jiangsu, China
| | | |
Collapse
|
47
|
Zhao Z, Wang Y, Peng J, Ren Z, Zhang L, Guo Q, Xu W, Lu S. Improvement in nerve regeneration through a decellularized nerve graft by supplementation with bone marrow stromal cells in fibrin. Cell Transplant 2012; 23:97-110. [PMID: 23128095 DOI: 10.3727/096368912x658845] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acellular nerve grafting is often inferior as well as an inadequate alternative to autografting for the repair of long gaps in peripheral nerves. Moreover, the injection method is not perfect. During the injection of cells, the syringe can destroy the acellular nerve structure and the limited accumulation of seed cells. To resolve this problem, we constructed a nerve graft by acellular nerve grafting. Bone marrow-mesenchymal stromal cells (BM-MSCs) were affixed with fibrin glue and injected inside or around the graft, which was then used to repair a 15-mm nerve defect in rats. The acellular nerve graft maintained its structure and composition, and its tensile strength was decreased, as determined by two-photon microscopy and a tensile testing device. In vitro, MSCs embedded in fibrin glue survived and secreted growth factors such as nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). We repaired 15-mm Sprague-Dawley rat sciatic nerve defects using this nerve graft construction, and MSCs injected around the graft helped improve nerve regeneration and functional recovery of peripheral nerve lesions as determined by functional analysis and histology. Therefore, we conclude that supplying MSCs in fibrin glue around acellular nerves is successful in maintaining the nerve structure and can support nerve regeneration similar to the direct injection of MSCs into the acellular nerve for long nerve defects but may avoid destroying the nerve graft. The technique is simple and is another option for stem cell transplantation.
Collapse
|
48
|
Wang Y, Jia H, Li WY, Tong XJ, Liu GB, Kang SW. Synergistic effects of bone mesenchymal stem cells and chondroitinase ABC on nerve regeneration after acellular nerve allograft in rats. Cell Mol Neurobiol 2012; 32:361-71. [PMID: 22095068 DOI: 10.1007/s10571-011-9764-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/11/2011] [Indexed: 12/23/2022]
Abstract
This study aimed to evaluate whether combination therapy of bone marrow stromal cells (BMSCs) transplantation and chondroitinase ABC (ChABC) treatment further enhances axonal regeneration and functional recovery after acellular nerve allograft repair of the sciatic nerve gap in rats. Eight Sprague-Dawley rats were used as nerve donors, and 32 Wistar rats were randomly divided into four groups: Group I: acellular rat sciatic nerve (ARSN) group; Group II: ChABC treatment; Group III: BMSCs transplantation; and Group IV: ChABC treatment and BMSCs transplantation. The results showed that compared with ARSN control group, BMSC transplantation promoted axonal regeneration, the secretion of neural trophic factors NGF, BDNF and axon angiogenesis in nerve graft. ChABC treatment degraded chondroitin sulfate proteoglycans in ARSN in vitro and in vivo and improved BMSCs survival in ARSN. The combination therapy caused much better beneficial effects evidenced by increasing sciatic function index, nerve conduction velocity, restoration rate of tibialis anterior wet muscle weight, and myelinated nerve number, but did not further boost the therapeutic effects on neurotrophic factor production, axon angiogenesis, and sensory functional recovery by BMSC transplantation. Taken together, for the first time, we demonstrate the synergistic effects of BMSC transplantation and BMSCs treatment on peripheral nerve regeneration, and our findings may help establish novel strategies for cell transplantation therapy for peripheral nerve injury.
Collapse
Affiliation(s)
- Ying Wang
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
49
|
Kvist M, Sondell M, Kanje M, Dahlin LB. Regeneration in, and properties of, extracted peripheral nerve allografts and xenografts. J Plast Surg Hand Surg 2011; 45:122-8. [PMID: 21682608 DOI: 10.3109/2000656x.2011.571847] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
When not enough conventional autologous nerve grafts are available, alternatives are needed to bridge nerve defects. Our aim was to study regeneration of nerves in chemically-extracted acellular nerve grafts from frogs, mice, humans (fresh and stored sural nerve), pigs and rats when defects in rat sciatic nerves were bridged. Secondly, we compared two different extraction procedures (techniques described by Sondell et al. and Hudson et al.) with respect to how efficiently they supported axonal outgrowth, and remaining laminin and myelin basic protein (MBP), after extraction. Isografts (rat) and xenografts (mouse) were transplanted into defects in rat sciatic nerves. Acellular nerve allografts from rats, extracted by the Sondell et al's technique, had an appreciably longer axonal outgrowth based on immunohistochemical staining of neurofilaments, than acellular nerve xenografts except those from the pig. Among acellular xenografts there was considerably longer axonal outgrowth in the grafts from pigs compared with those from humans (fresh), but there were no other differences among the xenografts with respect to axonal outgrowth. Axonal outgrowth in acellular nerve xenografts from mice, extracted by the method described by Sondell et al. was longer than in those extracted by Hudson et al's method, while there was no difference in outgrowth between extracted nerve isografts from rats. Electrophoretic analysis of extracted acellular nerve grafts showed remaining laminin, but not MBP, after both extraction procedures. These preserved laminin and removed MBP in acellular nerve grafts. Such grafts can be used to reconstruct short defects in nerves irrespective of their origin. However, selecting and matching a suitable combination of graft and host species may improve axonal outgrowth.
Collapse
Affiliation(s)
- Martin Kvist
- Department of Clinical Sciences Malmö, Hand Surgery, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | | | | |
Collapse
|
50
|
Jia H, Wang Y, Tong XJ, Liu GB, Li Q, Zhang LX, Sun XH. Biocompatibility of acellular nerves of different mammalian species for nerve tissue engineering. ACTA ACUST UNITED AC 2011; 39:366-75. [PMID: 21999105 DOI: 10.3109/10731199.2011.618133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
To explore the biocompatibility of acellular nerves of different mammalian species, for the acellular nerves derived from rats and rabbits, the morphology, immunocompatibility, and cytocompatibility with bone marrow stromal cells (BMSCs) were evaluated. The results indicated that the tridimensional architecture and main proteins of endoneurial tubes in both biomaterials were well retained. The nerve scaffolds did not show immunogenicity or cytotoxicity, but facilitated growth of BMSCs and secretion of neurotrophic factors in vitro. In conclusion, acellular nerves of different species possess favorable biocompatibility, and xenogenic acellular nerves combined with BMSCs have potential to replace allografts for peripheral nerve reconstruction.
Collapse
Affiliation(s)
- Hua Jia
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | | | | | | | | | | | | |
Collapse
|