1
|
Ajekiigbe VO, Agbo CE, Ogieuhi IJ, Anthony CS, Adewole OA, Ahmed B, Akingbola A, Nwankwo CK, Kayode AT, Chima UE, Adaobi OM. Innovative approaches to treatment of eye diseases: advances in stem cell therapy use in ophthalmology. Int Ophthalmol 2025; 45:113. [PMID: 40120030 DOI: 10.1007/s10792-025-03493-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION The human eye, a photo-sensory organ with an array of neuronal and tissue networks, remains susceptible to damage from various diseases and disorders despite its being a flawless masterpiece. It is estimated that over 2 billion people suffer from vision loss with common causative factors such as; age-related macular degeneration (AMD), glaucoma, cataracts, diabetic retinopathy, and infections amongst others. The use of Orthodox procedures has only helped mitigate the pathology; however, it doesn't serve any substantial curative purpose. More recently, the incorporation of new therapies via ocular delivery of nanomaterials and stem cell intervention has helped to change tides in the treatment of various ophthalmic pathologies. MAIN TEXT This review provides an overview of the current trends and breakthroughs in ophthalmology via stem cell therapy, with emphasis on its types, mechanisms, applications, and benefits. Mesenchymal stem cells which can arise from embryonic or adult origin possess some immunomodulatory effects that contribute to the therapeutic relevance of the MSCs and the ability to evade rejection from the host. However, the major drawback has been uncontrolled growth which can result in unintended side effects. Moreso, religious and ethical issues concerning the employment of MSCs from embryonic origin have also hindered clinical progression with its use. The use of stem cell therapy in the treatment of eye pathologies which is still undergoing clinical trials has shown to be a more viable treatment approach in ophthalmology as it targets retinal degenerative diseases thereby offering novel pathways for vision restoration. And also serves as a revolutionary alternative for treating severe ocular diseases. Stem cell delivery techniques might be quite cumbersome as the eye is a very delicate organ. The therapeutic interventional technique employed is aimed to ensure the reduction or absence of undesired effects in the deposition of the active pharmaceutical ingredient (API) being the stem cells. Techniques such as hydrogel-based injectables, which offer delivery of the APIs to the desired site of action without change in the physicochemical properties of the drug molecule, the scaffold delivery techniques, and the use of 3D bio-printing which can be used to develop scaffolds for retinal degeneration. The employment of artificial intelligence and machine learning in stem cell therapy has shown to be very fast and efficient in stem cell delivery and preventing likely human errors. CONCLUSIONS Unlike conventional treatments that often focus on managing symptoms, stem cells have the unique ability to repair and regenerate damaged tissues, addressing the root causes of the diseases. However, limitations due to economic, regulatory, and ethical challenges have posed barriers to advancing stem cell therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Bisharat Ahmed
- Allama Iqbal Medical College, University of Health Sciences, Lahore, Pakistan
| | - Adewunmi Akingbola
- Department of Public Health, University of Cambridge Cambridgeshire Old Trinity Schools, Cambridge, CB2 1TN, England, UK
| | | | | | | | | |
Collapse
|
2
|
Olsen TC, LaGuardia JS, Chen DR, Lebens RS, Huang KX, Milek D, Noble M, Leckenby JI. Influencing factors and repair advancements in rodent models of peripheral nerve regeneration. Regen Med 2024; 19:561-577. [PMID: 39469920 PMCID: PMC11633413 DOI: 10.1080/17460751.2024.2405318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/12/2024] [Indexed: 10/30/2024] Open
Abstract
Peripheral nerve injuries lead to severe functional impairments, with rodent models essential for studying regeneration. This review examines key factors affecting outcomes. Age-related declines, like reduced nerve fiber density and impaired axonal transport of vesicles, hinder recovery. Hormonal differences influence regeneration, with BDNF/trkB critical for testosterone and nerve growth factor for estrogen signaling pathways. Species and strain selection impact outcomes, with C57BL/6 mice and Sprague-Dawley rats exhibiting varying regenerative capacities. Injury models - crush for early regeneration, chronic constriction for neuropathic pain, stretch for traumatic elongation and transection for severe lacerations - provide insights into clinically relevant scenarios. Repair techniques, such as nerve grafts and conduits, show that autografts are the gold standard for gaps over 3 cm, with success influenced by graft type and diameter. Time course analysis highlights crucial early degeneration and regeneration phases within the first month, with functional recovery stabilizing by three to six months. Early intervention optimizes regeneration by reducing scar tissue formation, while later interventions focus on remyelination. Understanding these factors is vital for designing robust preclinical studies and translating research into effective clinical treatments for peripheral nerve injuries.
Collapse
Affiliation(s)
- Timothy C Olsen
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - Jonnby S LaGuardia
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - David R Chen
- University of California, 410 Charles E. Young Drive, East Los Angeles, CA90095, USA
| | - Ryan S Lebens
- University of California, 410 Charles E. Young Drive, East Los Angeles, CA90095, USA
| | - Kelly X Huang
- University of California, 410 Charles E. Young Drive, East Los Angeles, CA90095, USA
| | - David Milek
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - Mark Noble
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| | - Jonathan I Leckenby
- Division of Plastic & Reconstructive Surgery, University of Rochester Medical Center, 601 Elmwood Avenue Box 661Rochester, NY14642, USA
| |
Collapse
|
3
|
Liu T, Xu J, Pan X, Ding Z, Xie H, Wang X, Xie H. Advances of adipose-derived mesenchymal stem cells-based biomaterial scaffolds for oral and maxillofacial tissue engineering. Bioact Mater 2021; 6:2467-2478. [PMID: 33553828 PMCID: PMC7850942 DOI: 10.1016/j.bioactmat.2021.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/03/2021] [Accepted: 01/11/2021] [Indexed: 02/05/2023] Open
Abstract
The management of oral and maxillofacial tissue defects caused by tumors, trauma, and congenital or acquired deformities has been a major challenge for surgeons over the last few decades. Autologous tissue transplantation, the gold standard of tissue reconstruction, is a valid method for repairing the oral and maxillofacial functions and aesthetics. However, several limitations hinder its clinical applications including complications of donor sites, limited tissue volume, and uncertain long-term outcomes. Adipose-derived mesenchymal stem cells (ADMSCs) widely exist in adipose tissue and can be easily obtained through liposuction. Like the bone marrow-derived mesenchymal stem cells (BMSCs), ADMSCs also have the multi-pluripotent potencies to differentiate into osteoblasts, chondrocytes, neurons, and myocytes. Therefore, the multilineage capacity of ADMSCs makes them valuable for cell-based medical therapies. In recent years, researchers have developed many candidates of ADMSCs-based biomaterial scaffolds to cater for the needs of oral and maxillofacial tissue engineering due to their superior performance. This review presents the advances and applications of ADMSCs-based biomaterial scaffolds, and explores their tissue engineering prospects in oral and maxillofacial reconstructions.
Collapse
Affiliation(s)
- Tong Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jia Xu
- The Key Laboratory of Oral Biomedicine, Jiangxi Province, School of Stomatology, Nanchang University, Nanchang, 330006, China
| | - Xun Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhangfan Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hao Xie
- General Surgery Department, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241000, China
| | - Xiaoyi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Yoo MC, Chon J, Jung J, Kim SS, Bae S, Kim SH, Yeo SG. Potential Therapeutic Strategies and Substances for Facial Nerve Regeneration Based on Preclinical Studies. Int J Mol Sci 2021; 22:ijms22094926. [PMID: 34066483 PMCID: PMC8124575 DOI: 10.3390/ijms22094926] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Despite advances in microsurgical technology and an improved understanding of nerve regeneration, obtaining satisfactory results after facial nerve injury remains a difficult clinical problem. Among existing peripheral nerve regeneration studies, relatively few have focused on the facial nerve, particularly how experimental studies of the facial nerve using animal models play an essential role in understanding functional outcomes and how such studies can lead to improved axon regeneration after nerve injury. The purpose of this article is to review current perspectives on strategies for applying potential therapeutic methods for facial nerve regeneration. To this end, we searched Embase, PubMed, and the Cochrane library using keywords, and after applying exclusion criteria, obtained a total of 31 qualifying experimental studies. We then summarize the fundamental experimental studies on facial nerve regeneration, highlighting recent bioengineering studies employing various strategies for supporting facial nerve regeneration, including nerve conduits with stem cells, neurotrophic factors, and/or other therapeutics. Our summary of the methods and results of these previous reports reveal a common feature among studies, showing that various neurotrophic factors arising from injured nerves contribute to a microenvironment that plays an important role in functional recovery. In most cases, histological examinations showed that this microenvironmental influence increased axonal diameter as well as myelination thickness. Such an analysis of available research on facial nerve injury and regeneration represents the first step toward future therapeutic strategies.
Collapse
Affiliation(s)
- Myung Chul Yoo
- Department of Physical Medicine & Rehabilitation, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (M.C.Y.); (J.C.)
| | - Jinmann Chon
- Department of Physical Medicine & Rehabilitation, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (M.C.Y.); (J.C.)
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Sung Su Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Seonhwan Bae
- Department of Otorhinolaryngology, Head & Neck Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (S.B.); (S.H.K.)
| | - Sang Hoon Kim
- Department of Otorhinolaryngology, Head & Neck Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (S.B.); (S.H.K.)
| | - Seung Geun Yeo
- Department of Otorhinolaryngology, Head & Neck Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea; (S.B.); (S.H.K.)
- Correspondence: ; Tel.: +82-2-958-8980; Fax: +82-2-958-8470
| |
Collapse
|
5
|
Hopf A, Schaefer DJ, Kalbermatten DF, Guzman R, Madduri S. Schwann Cell-Like Cells: Origin and Usability for Repair and Regeneration of the Peripheral and Central Nervous System. Cells 2020; 9:E1990. [PMID: 32872454 PMCID: PMC7565191 DOI: 10.3390/cells9091990] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/06/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
Functional recovery after neurotmesis, a complete transection of the nerve fiber, is often poor and requires a surgical procedure. Especially for longer gaps (>3 mm), end-to-end suturing of the proximal to the distal part is not possible, thus requiring nerve graft implantation. Artificial nerve grafts, i.e., hollow fibers, hydrogels, chitosan, collagen conduits, and decellularized scaffolds hold promise provided that these structures are populated with Schwann cells (SC) that are widely accepted to promote peripheral and spinal cord regeneration. However, these cells must be collected from the healthy peripheral nerves, resulting in significant time delay for treatment and undesired morbidities for the donors. Therefore, there is a clear need to explore the viable source of cells with a regenerative potential similar to SC. For this, we analyzed the literature for the generation of Schwann cell-like cells (SCLC) from stem cells of different origins (i.e., mesenchymal stem cells, pluripotent stem cells, and genetically programmed somatic cells) and compared their biological performance to promote axonal regeneration. Thus, the present review accounts for current developments in the field of SCLC differentiation, their applications in peripheral and central nervous system injury, and provides insights for future strategies.
Collapse
Affiliation(s)
- Alois Hopf
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
| | - Dirk J. Schaefer
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Daniel F. Kalbermatten
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Neurosurgery, University Hospital Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| | - Srinivas Madduri
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland; (A.H.); (D.F.K.)
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland; (D.J.S.); (R.G.)
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, Spitalstrasse 21, 4031 Basel, Switzerland
| |
Collapse
|
6
|
Katiyar KS, Struzyna LA, Morand JP, Burrell JC, Clements B, Laimo FA, Browne KD, Kohn J, Ali Z, Ledebur HC, Smith DH, Cullen DK. Tissue Engineered Axon Tracts Serve as Living Scaffolds to Accelerate Axonal Regeneration and Functional Recovery Following Peripheral Nerve Injury in Rats. Front Bioeng Biotechnol 2020; 8:492. [PMID: 32523945 PMCID: PMC7261940 DOI: 10.3389/fbioe.2020.00492] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/28/2020] [Indexed: 12/23/2022] Open
Abstract
Strategies to accelerate the rate of axon regeneration would improve functional recovery following peripheral nerve injury, in particular for cases involving segmental nerve defects. We are advancing tissue engineered nerve grafts (TENGs) comprised of long, aligned, centimeter-scale axon tracts developed by the controlled process of axon "stretch-growth" in custom mechanobioreactors. The current study used a rat sciatic nerve model to investigate the mechanisms of axon regeneration across nerve gaps bridged by TENGs as well as the extent of functional recovery compared to nerve guidance tubes (NGT) or autografts. We established that host axon growth occurred directly along TENG axons, which mimicked the action of "pioneer" axons during development by providing directed cues for accelerated outgrowth. Indeed, axon regeneration rates across TENGs were 3-4 fold faster than NGTs and equivalent to autografts. The infiltration of host Schwann cells - traditional drivers of peripheral axon regeneration - was also accelerated and progressed directly along TENG axons. Moreover, TENG repairs resulted in functional recovery levels equivalent to autografts, with both several-fold superior to NGTs. These findings demonstrate that engineered axon tracts serve as "living scaffolds" to guide host axon outgrowth by a new mechanism - which we term "axon-facilitated axon regeneration" - that leads to enhanced functional recovery.
Collapse
Affiliation(s)
- Kritika S. Katiyar
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Axonova Medical LLC, Philadelphia, PA, United States
| | - Laura A. Struzyna
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Joseph P. Morand
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Justin C. Burrell
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Basak Clements
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Franco A. Laimo
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Kevin D. Browne
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Zarina Ali
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Douglas H. Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Axonova Medical LLC, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Axonova Medical LLC, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Zhao D, Li YH, Yang ZY, Cai T, Wu XY, Xia Y, Zhou Z. [Effect of the local application of stem cells on repairing facial nerve defects: a systematic review]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:59-68. [PMID: 32037768 DOI: 10.7518/hxkq.2020.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
OBJECTIVE To systematically evaluate the repairing effect of stem cells on facial nerve defects. METHODS Articles regarding the regenerating effect of stem cells on facial nerves in animals were collected from the databases of Pubmed, Cochrane Library, Web of Science, Embase, Scopus, and CBM. Two professionals independently completed the article screening, data extraction, and bias risk assessment. RevMan 5.3 and random-effects models were used for the statistical analysis, and the results were presented in the form of mean differences (MD) with a 95%CI. The results of functional evaluation (vibrissae movement, facial paralysis) and histological evaluation (density of myelinated fibers, diameter of fibers, thickness of myelin sheath, G ratio) of facial nerve were Meta-analyzed. RESULTS A total of 4 614 articles were retrieved from the 6 databases, and 15 of these articles were included in the Meta-analysis. For vibrissae movement and facial paralysis, the stem cell group scored significantly higher than the non-stem cell group (P<0.05). The density of myelinated fibers and thickness of the myelin sheath in the stem cell group were higher than those in the non-stem cell group (P<0.05). The G ratio in the stem cell group was smaller than that in the non-stem cell group (P=0.001). There was no significant difference in fiber diameter (P=0.08). CONCLUSIONS Stem cells have potential in promoting facial nerve regeneration.
Collapse
Affiliation(s)
- Dan Zhao
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| | - Yue-Heng Li
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| | - Zheng-Yan Yang
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| | - Ting Cai
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| | - Xiao-Yan Wu
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| | - Yu Xia
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| | - Zhi Zhou
- Dept. of Preventive Stomatology, Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Muni-cipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401120, China
| |
Collapse
|
8
|
The Use and Delivery of Stem Cells in Nerve Regeneration: Preclinical Evidence and Regulatory Considerations. Ann Plast Surg 2019; 80:448-456. [PMID: 29166311 DOI: 10.1097/sap.0000000000001259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Outcomes following peripheral nerve injury remain poor despite the regenerative capacity displayed by the peripheral nervous system. Current therapies are limited and do not provide satisfactory functional recovery in a multitude of cases. Biomaterials have decreased the need for nerve autograft across small nerve gaps in small-caliber nerves, but the lack of a cellular substrate presents a limiting factor to the effectiveness of this therapy. Schwann cells are the supportive cells in the peripheral nervous system and play an integral role in the physiological response and regeneration following nerve injury. Limitations to autologous Schwann cells include donor site morbidity during harvesting, limited expansion capability, and finite source. Stem cells are multipotent or pluripotent cells with self-renewing capabilities that show promise to improve functional recovery following nerve injury. Differentiation of stem cells into supportive Schwann cells could provide additional trophic support without the disadvantages of autologous Schwann cells, providing an avenue to improve existing therapies. A variety of stem cells have been evaluated in animal models for this clinical application; the current options, along with their clinical feasibility, are summarized in this article.
Collapse
|
9
|
Gonzalez-Perez F, Hernández J, Heimann C, Phillips JB, Udina E, Navarro X. Schwann cells and mesenchymal stem cells in laminin- or fibronectin-aligned matrices and regeneration across a critical size defect of 15 mm in the rat sciatic nerve. J Neurosurg Spine 2018; 28:109-118. [DOI: 10.3171/2017.5.spine161100] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVEArtificial nerve guides are being developed to substitute for autograft repair after peripheral nerve injuries. However, the use of conduits is limited by the length of the gap that needs to be bridged, with the success of regeneration highly compromised in long gaps. Addition of aligned proregenerative cells and extracellular matrix (ECM) components inside the conduit can be a good strategy to achieve artificial grafts that recreate the natural environment offered by a nerve graft. The purpose of this study was to functionalize chitosan devices with different cell types to support regeneration in limiting gaps in the rat peripheral nerve.METHODSThe authors used chitosan devices combined with proteins of the ECM and cells in a rat model of sciatic nerve injury. Combinations of fibronectin and laminin with mesenchymal stem cells (MSCs) or Schwann cells (SCs) were aligned within tethered collagen-based gels, which were placed inside chitosan tubes that were then used to repair a critical-size gap of 15 mm in the rat sciatic nerve. Electrophysiology and algesimetry tests were performed to analyze functional recovery during the 4 months after injury and repair. Histological analysis was performed at the midlevel and distal level of the tubes to assess the number of regenerated myelinated fibers.RESULTSFunctional analysis demonstrated that SC-aligned scaffolds resulted in 100% regeneration success in a 15-mm nerve defect in this rat model. In contrast, animals that underwent repair with MSC-aligned constructs had only 90% regeneration success, and those implanted with acellular bridges had only 75% regeneration success.CONCLUSIONSThese results indicate that the combination of chitosan conduits with ECM-enriched cellular gels represents a good alternative to the use of autografts for repairing long nerve gaps.
Collapse
Affiliation(s)
- Francisco Gonzalez-Perez
- 1Institute of Neurosciences and Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, and CIBERNED, Bellaterra, Spain
| | - Joaquim Hernández
- 1Institute of Neurosciences and Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, and CIBERNED, Bellaterra, Spain
| | | | - James B. Phillips
- 3Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, United Kingdom
| | - Esther Udina
- 1Institute of Neurosciences and Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, and CIBERNED, Bellaterra, Spain
| | - Xavier Navarro
- 1Institute of Neurosciences and Department of Cell Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, and CIBERNED, Bellaterra, Spain
| |
Collapse
|
10
|
Guo J, Guo S, Wang Y, Yu Y. Promoting potential of adipose derived stem cells on peripheral nerve regeneration. Mol Med Rep 2017; 16:7297-7304. [PMID: 28944869 PMCID: PMC5865858 DOI: 10.3892/mmr.2017.7570] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/25/2017] [Indexed: 01/08/2023] Open
Abstract
The ultimate goal of treating peripheral nerve defects is reconstructing continuity of the nerve stumps to regain nerve conduction and functional recovery. Clinically, autologous nerve grafts and Schwann cell (SC) therapy have limitations, such as the need for secondary surgery, sacrifice of another nerve and donor site complication. Adipose derived stem cells (ADSCs) may promise to be ideal alternative cells of SCs. To explore the potential of ADSCs promoting peripheral nerve regeneration, the present study investigated the influences of ADSCs on proliferation and neurotrophic function of SCs using co-culture model in vitro. Western blot analysis, immunocytochemistry, a cell viability assay, reverse transcription-polymerase chain reaction (RT-PCR) and ELISA were applied for examining the interaction of ADSCs and SCs in a co-culture model in vitro. Western blot analysis and immunocytochemistry demonstrated that protein expression levels of glial filament acidic protein (GFAP) and S100 in ADSCs co-cultured with SCs for 14 days were significantly higher compared with cells cultured alone. Cell viability assay indicated that the cell viability of SCs co-cultured with ADSCs for 3, 4, 5, 6 and 7 days was significantly higher than those cultured alone. RT-PCR showed that expression levels of neurotrophic factors [nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF)] and extracellular matrix components [fibronectin (FN) and laminin (LN)] in SCs co-cultured with ADSCs for 14 days were significantly higher than those in SCs cultured alone. NGF, GDNF, FN and LN in the supernatants of co-culture system were significantly higher than cells cultured alone, as ELISA revealed. The results of this study suggested that the transplantation of ADSCs may have a promoting potential to the peripheral nerve regeneration as undifferentiated state.
Collapse
Affiliation(s)
- Jiayan Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shu Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuxin Wang
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yanqiu Yu
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
11
|
Chitosan nerve conduits seeded with autologous bone marrow mononuclear cells for 30 mm goat peroneal nerve defect. Sci Rep 2017; 7:44002. [PMID: 28287100 PMCID: PMC5347120 DOI: 10.1038/srep44002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 02/03/2017] [Indexed: 01/01/2023] Open
Abstract
In the current research, to find if the combination of chitosan nerve conduits seeded with autologous bone marrow mononuclear cells (BM-MNCs) can be used to bridge 30 mm long peroneal nerve defects in goats, 15 animals were separated into BM-MNC group (n = 5), vehicle group (n = 5), and autologous nerve graft group (n = 5). 12 months after the surgery, animals were evaluated by behavioral observation, magnetic resonance imaging tests, histomorphological and electrophysiological analysis. Results revealed that animals in BM-MNC group and autologous nerve graft group achieved fine functional recovery; magnetic resonance imaging tests and histomorphometry analysis showed that the nerve defect was bridged by myelinated nerve axons in those animals. No significant difference was found between the two groups concerning myelinated axon density, axon diameter, myelin sheath thickness and peroneal nerve action potential. Animals in vehicle group failed to achieve significant functional recovery. The results indicated that chitosan nerve conduits seeded with autologous bone marrow mononuclear cells have strong potential in bridging long peripheral nerve defects and could be applied in future clinical trials.
Collapse
|
12
|
Osteogenic Differentiation Capacity of In Vitro Cultured Human Skeletal Muscle for Expedited Bone Tissue Engineering. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8619385. [PMID: 28210626 PMCID: PMC5292195 DOI: 10.1155/2017/8619385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/15/2016] [Accepted: 12/26/2016] [Indexed: 11/18/2022]
Abstract
Expedited bone tissue engineering employs the biological stimuli to harness the intrinsic regenerative potential of skeletal muscle to trigger the reparative process in situ to improve or replace biological functions. When genetically modified with adenovirus mediated BMP2 gene transfer, muscle biopsies from animals have demonstrated success in regenerating bone within rat bony defects. However, it is uncertain whether the human adult skeletal muscle displays an osteogenic potential in vitro when a suitable biological trigger is applied. In present study, human skeletal muscle cultured in a standard osteogenic medium supplemented with dexamethasone demonstrated significant increase in alkaline phosphatase activity approximately 24-fold over control at 2-week time point. More interestingly, measurement of mRNA levels revealed the dramatic results for osteoblast transcripts of alkaline phosphatase, bone sialoproteins, transcription factor CBFA1, collagen type I, and osteocalcin. Calcified mineral deposits were demonstrated on superficial layers of muscle discs after an extended 8-week osteogenic induction. Taken together, these are the first data supporting human skeletal muscle tissue as a promising potential target for expedited bone regeneration, which of the technologies is a valuable method for tissue repair, being not only effective but also inexpensive and clinically expeditious.
Collapse
|
13
|
Abstract
Tissue engineering of Schwann cells (SCs) can serve a number of purposes, such as in vitro SC-related disease modeling, treatment of peripheral nerve diseases or peripheral nerve injury, and, potentially, treatment of CNS diseases. SCs can be generated from autologous stem cells in vitro by recapitulating the various stages of in vivo neural crest formation and SC differentiation. In this review, we survey the cellular and molecular mechanisms underlying these in vivo processes. We then focus on the current in vitro strategies for generating SCs from two sources of pluripotent stem cells, namely embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Different methods for SC engineering from ESCs and iPSCs are reviewed and suggestions are proposed for optimizing the existing protocols. Potential safety issues regarding the clinical application of iPSC-derived SCs are discussed as well. Lastly, we will address future aspects of SC engineering.
Collapse
|
14
|
Past, Present, and Future of Nerve Conduits in the Treatment of Peripheral Nerve Injury. BIOMED RESEARCH INTERNATIONAL 2015; 2015:237507. [PMID: 26491662 PMCID: PMC4600484 DOI: 10.1155/2015/237507] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/12/2015] [Accepted: 05/19/2015] [Indexed: 01/03/2023]
Abstract
With significant advances in the research and application of nerve conduits, they have been used to repair peripheral nerve injury for several decades. Nerve conduits range from biological tubes to synthetic tubes, and from nondegradable tubes to biodegradable tubes. Researchers have explored hollow tubes, tubes filled with scaffolds containing neurotrophic factors, and those seeded with Schwann cells or stem cells. The therapeutic effect of nerve conduits is improving with increasing choice of conduit material, new construction of conduits, and the inclusion of neurotrophic factors and support cells in the conduits. Improvements in functional outcomes are expected when these are optimized for use in clinical practice.
Collapse
|
15
|
Kajbafzadeh AM, Khorramirouz R, Akbarzadeh A, Sabetkish S, Sabetkish N, Saadat P, Tehrani M. A novel technique for simultaneous whole-body and multi-organ decellularization: umbilical artery catheterization as a perfusion-based method in a sheep foetus model. Int J Exp Pathol 2015; 96:116-32. [PMID: 26031202 DOI: 10.1111/iep.12124] [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] [Received: 09/12/2014] [Accepted: 02/09/2015] [Indexed: 01/19/2023] Open
Abstract
The aim of this study was to develop a method to generate multi-organ acellular matrices. Using a foetal sheep model have developed a method of systemic pulsatile perfusion via the umbilical artery which allows for simultaneous multi-organ decellularization. Twenty sheep foetuses were systemically perfused with Triton X-100 and sodium dodecyl sulphate. Following completion of the whole-body decellularization, multiple biopsy samples were taken from different parts of 21 organs to ascertain complete cell component removal in the preserved extracellular matrices. Both the natural and decellularized organs were subjected to several examinations. The samples were obtained from the skin, eye, ear, nose, throat, cardiovascular, respiratory, gastrointestinal, urinary, musculoskeletal, central nervous and peripheral nervous systems. The histological results depicted well-preserved extracellular matrix (ECM) integrity and intact vascular structures, without any evidence of residual cellular materials, in all decellularized bioscaffolds. Scanning electron microscope (SEM) and biochemical properties remained intact, similar to their age-matched native counterparts. Preservation of the collagen structure was evaluated by a hydroxyproline assay. Dense organs such as bone and muscle were also completely decellularized, with a preserved ECM structure. Thus, as shown in this study, several organs and different tissues were decellularized using a perfusion-based method, which has not been previously accomplished. Given the technical challenges that exist for the efficient generation of biological scaffolds, the current results may pave the way for obtaining a variety of decellularized scaffolds from a single donor. In this study, there have been unique responses to the single acellularization protocol in foetuses, which may reflect the homogeneity of tissues and organs in the developing foetal body.
Collapse
Affiliation(s)
- Abdol-Mohammad Kajbafzadeh
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Reza Khorramirouz
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Aram Akbarzadeh
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Shabnam Sabetkish
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Nastaran Sabetkish
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Paria Saadat
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| | - Mona Tehrani
- Pediatric Urology Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran (IRI)
| |
Collapse
|
16
|
Sahin C, Karagoz H, Kulahci Y, Sever C, Akakin D, Kolbasi B, Ulkur E, Peker F. Minced nerve tissue in vein grafts used as conduits in rat tibial nerves. Ann Plast Surg 2015; 73:540-6. [PMID: 24691343 DOI: 10.1097/sap.0000000000000060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Peripheral nerve injuries are encountered frequently in clinical practice. In nerve repair, an end-to-end suture is the preferable choice of treatment. However, where primary closure is not possible, the defect is to be repaired with a nerve graft. METHODS A total of 21 female Wistar rats weighing 230 to 290 g were used in the study. They were classified into the following 3 groups: (I) nerve graft, (II) vein graft, and (III) minced nerve graft. In group I, after exposure of the tibial nerve, a 1-cm-long nerve gap was created on the tibial nerve, and the defect was repaired epineurally by using the autogenous nerve. In group II, the 1-cm tibial nerve defect was repaired by using an autogenous vein graft. In group III, a 1-cm nerve graft was divided to 3 equal parts, with one of the nerve parts being minced with microscissors and placed in the vein graft lumen. Thereafter, a 1-cm tibial nerve defect was repaired by the vein graft filled with minced nerve tissue. The tibial function indices (TFIs) were calculated for functional assessment using the Bain-Mackinnon-Hunter formula. Light and electron microscopic evaluations were performed for morphometric assessment. In addition, the myelinated fibers were counted in all groups. RESULTS The TFIs of group II were found to be the lowest among all the groups after the sixth week, whereas the TFI of group I was found to be better than the other groups after the sixth week. There was no difference in TFIs between group I and group III. On the basis of the number of myelinated fibers, there was no statistically significant difference between group I and group III, whereas the difference was significant (P<0.05) between groups I/III and group II. Presence of peripheral nerves in light microscopic evaluation revealed normal characteristics of myelinated fibers in all groups. The myelinated axon profile was near normal in the nerve graft group in electron microscopic evaluation. However, there were more degenerated axons with disturbed contours and vacuolizations in the vein graft group compared to the minced nerve graft group. CONCLUSIONS We can conclude that using minced nerve tissue in vein grafts as a conduit increases the regeneration of nerves (almost like the nerve graft group) and it may not be caused by donor-site morbidity. It can be used in the repair of nerve defects instead of autogenous nerve grafts after further experimental evidence and clinical trials.
Collapse
Affiliation(s)
- Cihan Sahin
- From the *Department of Plastic and Reconstructive Surgery, Gulhane Military Medical Academy, Haydarpasa Training Hospital, Istanbul; †Department of Hand and Upper Extremity Surgery, Gulhane Military Medical Academy, Ankara; ‡Department of Histology and Embryology, Marmara University, Medical School; and §F&P Plastic Reconstructive and Aesthetic Surgery Center, Istanbul, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Epineural Tube Repair. Plast Reconstr Surg 2015. [DOI: 10.1007/978-1-4471-6335-0_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Ye F, Li H, Qiao G, Chen F, Tao H, Ji A, Hu Y. Platelet-rich plasma gel in combination with Schwann cells for repair of sciatic nerve injury. Neural Regen Res 2014; 7:2286-92. [PMID: 25538751 PMCID: PMC4268730 DOI: 10.3969/j.issn.1673-5374.2012.29.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 08/25/2012] [Indexed: 01/26/2023] Open
Abstract
Bone marrow mesenchymal stem cells were isolated from New Zealand white rabbits, culture-expanded and differentiated into Schwann cell-like cells. Autologous platelet-rich plasma and Schwann cell-like cells were mixed in suspension at a density of 1 × 10(6) cells/mL, prior to introduction into a poly (lactic-co-glycolic acid) conduit. Fabricated tissue-engineered nerves were implanted into rabbits to bridge 10 mm sciatic nerve defects (platelet-rich plasma group). Controls were established using fibrin as the seeding matrix for Schwann cell-like cells at identical density to construct tissue-engineered nerves (fibrin group). Twelve weeks after implantation, toluidine blue staining and scanning electron microscopy were used to demonstrate an increase in the number of regenerating nerve fibers and thickness of the myelin sheath in the platelet-rich plasma group compared with the fibrin group. Fluoro-gold retrograde labeling revealed that the number of Fluoro-gold-positive neurons in the dorsal root ganglion and the spinal cord anterior horn was greater in the platelet-rich plasma group than in the fibrin group. Electrophysiological examination confirmed that compound muscle action potential and nerve conduction velocity were superior in the platelet-rich plasma group compared with the fibrin group. These results indicate that autologous platelet-rich plasma gel can effectively serve as a seeding matrix for Schwann cell-like cells to construct tissue-engineered nerves to promote peripheral nerve regeneration.
Collapse
Affiliation(s)
- Fagang Ye
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| | - Haiyan Li
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| | - Guangxi Qiao
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| | - Feng Chen
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| | - Hao Tao
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| | - Aiyu Ji
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| | - Yanling Hu
- Department of Trauma Surgery, Affiliated Hospital of Medical College, Qingdao University, Qingdao 266000, Shandong Province, China
| |
Collapse
|
19
|
Liu H, Wen W, Hu M, Bi W, Chen L, Liu S, Chen P, Tan X. Chitosan conduits combined with nerve growth factor microspheres repair facial nerve defects. Neural Regen Res 2014; 8:3139-47. [PMID: 25206635 PMCID: PMC4158708 DOI: 10.3969/j.issn.1673-5374.2013.33.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 06/10/2013] [Indexed: 01/09/2023] Open
Abstract
Microspheres containing nerve growth factor for sustained release were prepared by a compound method, and implanted into chitosan conduits to repair 10-mm defects on the right buccal branches of the facial nerve in rabbits. In addition, chitosan conduits combined with nerve growth factor or normal saline, as well as autologous nerve, were used as controls. At 90 days post-surgery, the muscular atrophy on the right upper lip was more evident in the nerve growth factor and normal sa-line groups than in the nerve growth factor-microspheres and autologous nerve groups. physiological analysis revealed that the nerve conduction velocity and amplitude were significantly higher in the nerve growth factor-microspheres and autologous nerve groups than in the nerve growth factor and normal saline groups. Moreover, histological observation illustrated that the di-ameter, number, alignment and myelin sheath thickness of myelinated nerves derived from rabbits were higher in the nerve growth factor-microspheres and autologous nerve groups than in the nerve growth factor and normal saline groups. These findings indicate that chitosan nerve conduits bined with microspheres for sustained release of nerve growth factor can significantly improve facial nerve defect repair in rabbits.
Collapse
Affiliation(s)
- Huawei Liu
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Weisheng Wen
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Min Hu
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Wenting Bi
- Department of Stomatology, Second Hospital of Beijing Chaoyang District, Beijing 100026, China
| | - Lijie Chen
- Department of Stomatology, First Sanatorium of Qingdao, Jinan Military Area Command of Chinese PLA, Qingdao 266071, Shandong Province, China
| | - Sanxia Liu
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Peng Chen
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| | - Xinying Tan
- Institute of Stomatology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
20
|
Kador KE, Goldberg JL. Scaffolds and stem cells: delivery of cell transplants for retinal degenerations. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 7:459-470. [PMID: 23585772 DOI: 10.1586/eop.12.56] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Retinal degenerations and optic neuropathies often lead to death of photoreceptors or retinal ganglion cells, respectively. Stem cell therapies are showing promise for these diseases in preclinical models and are beginning to transition into human trials, but cell delivery and integration remain major challenges. Focusing on photoreceptor- and progenitor-directed approaches, in this article, the authors review how advances in tissue engineering and cell scaffold design are enhancing cell therapies for retinal degeneration.
Collapse
Affiliation(s)
- Karl E Kador
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, 1501 NW 10th Avenue, BRB 826, FL 33136, USA
| | | |
Collapse
|
21
|
Euler de Souza Lucena E, Guzen FP, Lopes de Paiva Cavalcanti JR, Galvão Barboza CA, Silva do Nascimento Júnior E, Cavalcante JDS. Experimental considerations concerning the use of stem cells and tissue engineering for facial nerve regeneration: a systematic review. J Oral Maxillofac Surg 2013; 72:1001-12. [PMID: 24480768 DOI: 10.1016/j.joms.2013.11.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE Peripheral nerve trauma results in functional loss in the innervated organ, and recovery without surgical intervention is rare. Many surgical techniques can be used for repair in experimental models. The authors investigated the source and delivery method of stem cells in experimental outcomes, seeking to clarify whether stem cells must be differentiated in the injured facial nerve and improve the regenerative process. MATERIALS AND METHODS The following key terms were used: nervous regeneration, nerve regeneration, facial nerve regeneration, stem cells, embryonic stem cells, fetal stem cells, adult stem cells, facial nerve, facial nerve trauma, and facial nerve traumatism. The search was restricted to experimental studies that applied stem cell therapy and tissue engineering for nerve repair. RESULTS Eight studies meeting the inclusion criteria were reviewed. Different sources of stem and precursor cells were explored (bone marrow mesenchymal stem cells, adipose-derived stem cells, dental pulp cells, and neural stem cells) for their potential application in the scenario of facial nerve injuries. Different material conduits (vases, collagen, and polyglycolic acid) were used as bridges. Immunochemistry and electrophysiology are the principal methods for analyzing regenerative effects. Although recent studies have shown that stem cells can act as a promising bridge for nerve repair, considerable optimization of these therapies will be required for their potential to be realized in a clinical setting. CONCLUSION Based on these studies, the use of stem cells derived from different sources presents promising results related to facial nerve regeneration and produces effective functional results. The use of tubes also optimizes nerve repair, thus promoting greater myelination and axonal growth of peripheral nerves.
Collapse
Affiliation(s)
- Eudes Euler de Souza Lucena
- Assistant Professor, Laboratory of Experimental Neurology, Health Science Center, State University of Rio Grande do Norte, Mossoró, RN, Brazil.
| | - Fausto Pierdoná Guzen
- Adjunct Professor, Laboratory of Experimental Neurology, Health Science Center, State University of Rio Grande do Norte, Mossoró, RN, Brazil
| | | | - Carlos Augusto Galvão Barboza
- Associate Professor, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Expedito Silva do Nascimento Júnior
- Adjunct Professor, Laboratory of Neuroanatomy, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jeferson de Sousa Cavalcante
- Associate Professor, Laboratory of Neuroanatomy, Department of Morphology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
22
|
Zavan B, Abatangelo G, Mazzoleni F, Bassetto F, Cortivo R, Vindigni V. New 3D hyaluronan-based scaffold forin vitroreconstruction of the rat sciatic nerve. Neurol Res 2013; 30:190-6. [DOI: 10.1179/174313208x281082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
23
|
Costa HJZR, Bento RF, Salomone R, Azzi-Nogueira D, Zanatta DB, Paulino Costa M, da Silva CF, Strauss BE, Haddad LA. Mesenchymal bone marrow stem cells within polyglycolic acid tube observed in vivo after six weeks enhance facial nerve regeneration. Brain Res 2013; 1510:10-21. [PMID: 23542586 DOI: 10.1016/j.brainres.2013.03.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/04/2013] [Accepted: 03/18/2013] [Indexed: 12/15/2022]
Abstract
Autografting is the gold-standard method for facial nerve repair with tissue loss. Its association with high-quality scaffolds and cell implants has disclosed distinct experimental outcomes. The aim of this study was to evaluate the functional and histological effects of bone marrow stem cells (BMSC) combined with polyglycolic acid tube (PGAt) in autografted rat facial nerves. After neurotmesis of the mandibular branch of the rat facial nerve, surgical repair consisted of nerve autografting (groups A-E) contained in pGAT (groups B-E), filled with basement membrane matrix (groups C-E) with undifferentiated BMSC (group D) or Schwann-like cells that had differentiated from BMSC (group E). Axon morphometrics and an objective compound muscle action potentials (CMAP) analysis were conducted. Immunofluorescence assays were carried out with Schwann cell marker S100 and anti-β-galactosidase to label exogenous cells. Six weeks after surgery, animals from either cell-containing group had mean CMAP amplitudes significantly higher than control groups. Differently from other groups, facial nerves with Schwann-like cell implants had mean axonal densities within reference values. This same group had the highest mean axonal diameter in distal segments. We observed expression of the reporter gene lacZ in nerve cells in the graft and distally from it in groups D and E. Group-E cells had lacZ coexpressed with S100. In conclusion, regeneration of the facial nerve was improved by BMSC within PGAt in rats, yet Schwann-like cells were associated with superior effects. Accordingly, groups D and E had BMSC integrated in neural tissue with maintenance of former cell phenotype for six weeks.
Collapse
|
24
|
Liu BS, Yang YC, Shen CC. Regenerative effect of adipose tissue-derived stem cells transplantation using nerve conduit therapy on sciatic nerve injury in rats. J Tissue Eng Regen Med 2012; 8:337-50. [PMID: 22552954 DOI: 10.1002/term.1523] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 09/16/2011] [Accepted: 03/09/2012] [Indexed: 01/13/2023]
Affiliation(s)
- Bai-Shuan Liu
- Department of Medical Imaging and Radiological Sciences; Central Taiwan University of Science and Technology; Taichung Taiwan Republic of China
| | - Yi-Chin Yang
- Department of Neurosurgery; Taichung Veterans General Hospital; Taichung Taiwan Republic of China
| | - Chiung-Chyi Shen
- Department of Neurosurgery; Taichung Veterans General Hospital; Taichung Taiwan Republic of China
- Faculty of Medicine, School of Medicine; National Yang-Ming University; Taipei Taiwan Republic of China
| |
Collapse
|
25
|
Early regenerative effects of NGF-transduced Schwann cells in peripheral nerve repair. Mol Cell Neurosci 2012; 50:103-12. [PMID: 22735691 DOI: 10.1016/j.mcn.2012.04.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 03/27/2012] [Accepted: 04/06/2012] [Indexed: 01/04/2023] Open
Abstract
Peripheral nerve injury leads to a rapid and robust increase in the synthesis of neurotrophins which guide and support regenerating axons. To further optimize neurotrophin supply at the earliest stages of regeneration, we over-expressed NGF in Schwann cells (SCs) by transducing these cells with a lentiviral vector encoding NGF (NGF-SCs). Transplantation of NGF-SCs in a rat sciatic nerve transection/repair model led to significant increase of NGF levels 2weeks after injury and correspondingly to substantial improvement in axonal regeneration. Numbers of NF200, ChAT and CGRP-positive axon profiles, as well as the gastrocnemius muscle weights, were significantly higher in the NGF-Schwann cell group compared to the animals that received control SCs transduced with a lentiviral vector encoding GFP (GFP-SCs). Comparison with other models of NGF application signifies the important role of this neurotrophin during the early stages of regeneration, and supports the importance of developing combined gene and cell therapy for peripheral nerve repair.
Collapse
|
26
|
Lin YC, Marra KG. Injectable systems and implantable conduits for peripheral nerve repair. Biomed Mater 2012; 7:024102. [DOI: 10.1088/1748-6041/7/2/024102] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
27
|
Shen CC, Yang YC, Liu BS. Peripheral nerve repair of transplanted undifferentiated adipose tissue-derived stem cells in a biodegradable reinforced nerve conduit. J Biomed Mater Res A 2011; 100:48-63. [PMID: 21972223 DOI: 10.1002/jbm.a.33227] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/04/2011] [Accepted: 08/16/2011] [Indexed: 12/12/2022]
Abstract
This study proposes a biodegradable nerve conduit containing genipin-cross-linked gelatin annexed with tricalcium phosphate ceramic particles (genipin-gelatin-tricalcium phosphate, GGT) in peripheral nerve regeneration. Firstly, cytotoxicity tests revealed that the GGT-extracts were not toxic, and promoted the proliferation and neuronal differentiation of adipose tissue-derived stem cells (ADSCs). Secondly, the GGT composite film effectively supported ADSCs attachment and growth. Additionally, the GGT substrate was biocompatible with the neonatal rat sciatic nerve and produced a beneficial effect on peripheral nerve repair through in vitro tissue culture. Finally, the experiments in this study confirmed the effectiveness of a GGT/ADSCs nerve conduit as a guidance channel for repairing a 10-mm gap in a rat sciatic nerve. Eight weeks after implantation, the mean recovery index of compound muscle action potentials (CMAPs) was significantly different between the GGT/ADSCs and autografts groups (p < 0.05), both of which were significantly superior to the GGT group (p < 0.05). Furthermore, walking track analysis also showed a significantly higher sciatic function index (SFI) score (p < 0.05) and better toe spreading development in the GGT/ADSCs group than in the autograft group. Histological observations and immunohistochemistry revealed that the morphology and distribution patterns of nerve fibers in the GGT/ADSCs nerve conduits were similar to those of the autografts. The GGT nerve conduit offers a better scaffold for the incorporation of seeding undifferentiated ADSCs, and opens a new avenue to replace autologous nerve grafts for the rapid regeneration of damaged peripheral nerve tissues and an improved approach to patient care.
Collapse
Affiliation(s)
- Chiung-Chyi Shen
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan, Republic of China
| | | | | |
Collapse
|
28
|
Peripheral Nerve Defect Repair With Epineural Tubes Supported With Bone Marrow Stromal Cells. Ann Plast Surg 2011; 67:73-84. [DOI: 10.1097/sap.0b013e318223c2db] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
29
|
Yang LM, Liu XL, Zhu QT, Zhang Y, Xi TF, Hu J, He CF, Jiang L. Human peripheral nerve-derived scaffold for tissue-engineered nerve grafts: Histology and biocompatibility analysis. J Biomed Mater Res B Appl Biomater 2010; 96:25-33. [DOI: 10.1002/jbm.b.31719] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
30
|
Abstract
Bridging nerve gaps with suitable grafts is a major clinical problem. The autologous nerve graft is considered to be the gold standard, providing the best functional results; however, donor site morbidity is still a major disadvantage. Various attempts have been made to overcome the problems of autologous nerve grafts with artificial nerve tubes, which are “ready-to-use” in almost every situation. A wide range of materials have been used in animal models but only few have been applied to date clinically, where biocompatibility is an inevitable prerequisite. This review gives an idea about artificial nerve tubes with special focus on their biocompatibility in animals and humans.
Collapse
Affiliation(s)
- Felix Stang
- Department of Plastic, Reconstructive and Hand Surgery, University of Luebeck, 23538 Luebeck, Germany
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-451-5002061; Fax: +49-451-5002190
| | - Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, University of Magdeburg, 39120 Magdeburg, Germany; E-Mail:
| | - Hisham Fansa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld-Mitte, 33604 Bielefeld, Germany; E-Mail:
| |
Collapse
|
31
|
Huang JH, Cullen DK, Browne KD, Groff R, Zhang J, Pfister BJ, Zager EL, Smith DH. Long-term survival and integration of transplanted engineered nervous tissue constructs promotes peripheral nerve regeneration. Tissue Eng Part A 2009; 15:1677-85. [PMID: 19231968 DOI: 10.1089/ten.tea.2008.0294] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although peripheral nerve injury is a common consequence of trauma or surgery, there are insufficient means for repair. In particular, there is a critical need for improved methods to facilitate regeneration of axons across major nerve lesions. Here, we engineered transplantable living nervous tissue constructs to provide a labeled pathway to guide host axonal regeneration. These constructs consisted of stretch-grown, longitudinally aligned living axonal tracts inserted into poly(glycolic acid) tubes. The constructs (allogenic) were transplanted to bridge an excised segment of sciatic nerve in the rat, and histological analyses were performed at 6 and 16 weeks posttransplantation to determine graft survival, integration, and host regeneration. At both time points, the transplanted constructs were found to have maintained their pretransplant geometry, with surviving clusters of graft neuronal somata at the extremities of the constructs spanned by tracts of axons. Throughout the transplanted region, there was an intertwining plexus of host and graft axons, suggesting that the transplanted axons mediated host axonal regeneration across the lesion. By 16 weeks posttransplant, extensive myelination of axons was observed throughout the transplant region. Further, graft neurons had extended axons beyond the margins of the transplanted region, penetrating into the host nerve. Notably, this survival and integration of the allogenic constructs occurred in the absence of immunosuppression therapy. These findings demonstrate the promise of living tissue-engineered axonal constructs to bridge major nerve lesions and promote host regeneration, potentially by providing axon-mediated axonal outgrowth and guidance.
Collapse
Affiliation(s)
- Jason H Huang
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Walsh S, Biernaskie J, Kemp SWP, Midha R. Supplementation of acellular nerve grafts with skin derived precursor cells promotes peripheral nerve regeneration. Neuroscience 2009; 164:1097-107. [PMID: 19737602 DOI: 10.1016/j.neuroscience.2009.08.072] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 08/04/2009] [Accepted: 08/28/2009] [Indexed: 12/17/2022]
Abstract
Introduction of autologous stem cells into the site of a nerve injury presents a promising therapy to promote axonal regeneration and remyelination following peripheral nerve damage. Given their documented ability to differentiate into Schwann cells (SCs) in vitro, we hypothesized that skin-derived precursor cells (SKPs) could represent a clinically-relevant source of transplantable cells that would enhance nerve regeneration following peripheral nerve injury. In this study, we examined the potential for SKP-derived Schwann cells (SKP-SCs) or nerve-derived SCs to improve nerve regeneration across a 12 mm gap created in the sciatic nerve of Lewis rats bridged by a freeze-thawed nerve graft. Immunohistology after 4 weeks showed survival of both cell types and early regeneration in SKP seeded grafts was comparable to those seeded with SCs. Histomorphometrical and electrophysiological measurements of cell-treated nerve segments after 8 weeks survival all showed significant improvement as compared to diluent controls. A possible mechanistic explanation for the observed results of improved regenerative outcomes lies in SKP-SCs' ability to secrete bioactive neurotrophins. We therefore conclude that SKPs represent an easily accessible, autologous source of stem cells for transplantation therapies which act as functional Schwann cells and show great promise in improving regeneration following nerve injury.
Collapse
Affiliation(s)
- S Walsh
- Department of Clinical Neuroscience and Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Heritage Medical Research Building 109-3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| | | | | | | |
Collapse
|
33
|
Dinh P, Hazel A, Palispis W, Suryadevara S, Gupta R. Functional assessment after sciatic nerve injury in a rat model. Microsurgery 2009; 29:644-9. [DOI: 10.1002/micr.20685] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
Ma MS, Van Dam G, Meek M, Boddeke E, Copray S. In vivo bioluminescent imaging of Schwann cells in a poly(DL
-lactide-ε-caprolactone) nerve guide. Muscle Nerve 2009; 40:867-71. [DOI: 10.1002/mus.21372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Ishikawa N, Suzuki Y, Dezawa M, Kataoka K, Ohta M, Cho H, Ide C. Peripheral nerve regeneration by transplantation of BMSC-derived Schwann cells as chitosan gel sponge scaffolds. J Biomed Mater Res A 2009; 89:1118-24. [DOI: 10.1002/jbm.a.32389] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Walsh SK, Gordon T, Addas BMJ, Kemp SWP, Midha R. Skin-derived precursor cells enhance peripheral nerve regeneration following chronic denervation. Exp Neurol 2009; 223:221-8. [PMID: 19477174 DOI: 10.1016/j.expneurol.2009.05.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 05/13/2009] [Accepted: 05/18/2009] [Indexed: 12/23/2022]
Abstract
While peripheral nerves demonstrate the capacity for axonal regeneration, outcome following injury remains relatively poor, especially following prolonged denervation. Since axon-deprived Schwann cells (SCs) in the distal nerve progressively lose their ability to support axonal growth, we took the approach of using skin-derived precursor cells (SKPs) as an accessible source of replacement SCs that could be transplanted into chronically denervated peripheral nerve. In this study, we employed a delayed cross-reinnervation paradigm to assess regeneration of common peroneal nerve axons into the chronically denervated rodent tibial nerve following delivery of SKP-derived SC (SKP-SCs). SKP-SC treated animals exhibited superior axonal regeneration to media controls, with significantly higher counts of regenerated motorneurons and histological recovery similar to that of immediately repaired nerve. Improved axonal regeneration correlated with superior muscle reinnervation, as measured by compound muscle action potentials and wet muscle weights. We therefore conclude that SKPs represent an easily accessible, autologous source of stem cell-derived Schwann cells that show promise in improving regeneration through chronically injured nerves.
Collapse
Affiliation(s)
- Sarah K Walsh
- Department of Clinical Neuroscience and Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, HMRB 109-3330 Hospital Drive NW, Calgary, Alberta, Canada T2N4N1.
| | | | | | | | | |
Collapse
|
37
|
Stern MM, Myers RL, Hammam N, Stern KA, Eberli D, Kritchevsky SB, Soker S, Van Dyke M. The influence of extracellular matrix derived from skeletal muscle tissue on the proliferation and differentiation of myogenic progenitor cells ex vivo. Biomaterials 2009; 30:2393-9. [PMID: 19168212 PMCID: PMC9850446 DOI: 10.1016/j.biomaterials.2008.12.069] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Accepted: 12/28/2008] [Indexed: 01/21/2023]
Abstract
Skeletal muscle relies upon regeneration to maintain homeostasis and repair injury. This process involves the recruitment of the tissue's resident stem cell, the muscle progenitor cell, and a subsequent proliferative response by newly generated myoblasts, which must then align and fuse to generate new muscle fibers. During regeneration, cells rely on environmental input for direction. Extracellular matrix (ECM) represents a crucial component of a cell's microenvironment that aids in guiding muscle regeneration. We hypothesized that ECM extracted from skeletal muscle would provide muscle progenitor cells and myoblasts with an ideal substrate for growth and differentiation ex vivo. To test this hypothesis, we developed a method to extract ECM from the large thigh muscles of adult rats and present it to cells as a surface coating. Myogenic cells cultured on ECM extract experienced enhanced proliferation and differentiation relative to standard growth surfaces. As the methodology can be applied to any size muscle, these results demonstrate that bioactive ECM can be readily obtained from skeletal muscle and used to develop biomaterials that enhance muscle regeneration. Furthermore, the model system demonstrated here can be applied to the study of interactions between the ECM of a particular tissue and a cell population of interest.
Collapse
Affiliation(s)
- Matthew M. Stern
- J. Paul Sticht Center on Aging and Rehabilitation, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA,Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Regina L. Myers
- J. Paul Sticht Center on Aging and Rehabilitation, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA,Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Nevin Hammam
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Kathryn A. Stern
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Daniel Eberli
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Stephen B. Kritchevsky
- J. Paul Sticht Center on Aging and Rehabilitation, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Mark Van Dyke
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA,Corresponding author. Tel.: +1 336 713 7266; fax: +1 336 713 7290. (M. Van Dyke)
| |
Collapse
|
38
|
Application of neural stem cells in tissue-engineered artificial nerve. Otolaryngol Head Neck Surg 2009; 140:159-64. [PMID: 19201281 DOI: 10.1016/j.otohns.2008.10.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2008] [Revised: 10/27/2008] [Accepted: 10/27/2008] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To observe the curative effect of neural stem cells (NSCs), which are used in tissue-engineered artificial nerve, on repairing rabbit 10-mm facial nerve defects. METHODS Thirty-six Oryctolagus cuniculi were randomly divided into three groups (each group with 12 Oryctolagus cuniculi). In group A, chitosan conduit, collagen protein sponge, nerve growth factor (NGF), and NSCs were used. In group B, chitosan conduit, collagen sponge, and NGF were used. In group C, nerve autograft was performed. Electrophysiologic detection, histologic observation, and BrdU and S100 immunohistochemical examination were performed 12 weeks after operation. RESULTS All observation items in group A were better than those in group B (P < 0.01), and there were no significant differences between group A and group C (P > 0.05). CONCLUSION NSCs may be served as seed cells of peripheral nerve tissue engineering and be used in artificial nerve to repair facial nerve defects.
Collapse
|
39
|
|
40
|
Zhang H, Wei YT, Tsang KS, Sun CR, Li J, Huang H, Cui FZ, An YH. Implantation of neural stem cells embedded in hyaluronic acid and collagen composite conduit promotes regeneration in a rabbit facial nerve injury model. J Transl Med 2008; 6:67. [PMID: 18986538 PMCID: PMC2614414 DOI: 10.1186/1479-5876-6-67] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2008] [Accepted: 11/05/2008] [Indexed: 12/14/2022] Open
Abstract
The implantation of neural stem cells (NSCs) in artificial scaffolds for peripheral nerve injuries draws much attention. NSCs were ex-vivo expanded in hyaluronic acid (HA)-collagen composite with neurotrophin-3, and BrdU-labeled NSCs conduit was implanted onto the ends of the transected facial nerve of rabbits. Electromyography demonstrated a progressive decrease of current threshold and increase of voltage amplitude in de-innervated rabbits after implantation for one, four, eight and 12 weeks compared to readouts derived from animals prior to nerve transection. The most remarkable improvement, observed using Electrophysiology, was of de-innervated rabbits implanted with NSCs conduit as opposed to de-innervated counterparts with and without the implantation of HA-collagen, NSCs and HA-collagen, and HA-collagen and neurotrophin-3. Histological examination displayed no nerve fiber in tissue sections of de-innervated rabbits. The arrangement and S-100 immunoreactivity of nerve fibers in the tissue sections of normal rabbits and injured rabbits after implantation of NSCs scaffold for 12 weeks were similar, whereas disorderly arranged minifascicles of various sizes were noted in the other three arms. BrdU+ cells were detected at 12 weeks post-implantation. Data suggested that NSCs embedded in HA-collagen biomaterial could facilitate re-innervations of damaged facial nerve and the artificial conduit of NSCs might offer a potential treatment modality to peripheral nerve injuries.
Collapse
Affiliation(s)
- Han Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sakimoto T, Kim TI, Ellenberg D, Fukai N, Jain S, Azar DT, Chang JH. Collagen XVIII and corneal reinnervation following keratectomy. FEBS Lett 2008; 582:3674-80. [PMID: 18840438 DOI: 10.1016/j.febslet.2008.09.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 09/14/2008] [Accepted: 09/25/2008] [Indexed: 10/21/2022]
Abstract
The significance of collagen XVIII in the regulation of corneal reinnervation remains largely unknown. We used whole-mount immunoconfocal microscopy to localize collagen XVIII to the nerve basement membrane of wild-type (WT) mouse corneas. Transmission electron microscopy showed corneal nerve disorganization in collagen XVIII knockout mice (col18a1(-/-)). Antibody 2H3-specific neurofilament colocalized with collagens XVIII and IV and laminin-2 in WT mouse corneas, but did not colocalize with collagen IV and laminin-2 in col18a1(-/-) mouse corneas. Following keratectomy, col18a1(-/-) mice displayed decreased corneal neurite extension compared to WT mice. Our data indicate that collagen XVIII may play an important role in corneal reinnervation after wounding.
Collapse
Affiliation(s)
- Tohru Sakimoto
- Schepens Eye Research Institute, Harvard University, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Keilhoff G, Schild L, Fansa H. Minocycline protects Schwann cells from ischemia-like injury and promotes axonal outgrowth in bioartificial nerve grafts lacking Wallerian degeneration. Exp Neurol 2008; 212:189-200. [PMID: 18501894 DOI: 10.1016/j.expneurol.2008.03.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 03/25/2008] [Accepted: 03/26/2008] [Indexed: 01/08/2023]
Abstract
Minocycline, a broad-spectrum antimicrobial tetracycline, acts neuroprotectively in ischemia. Recently, however, minocycline has been revealed to have ambiguous effects on nerve regeneration. Thus its effects in a rat sciatic nerve transplantation model and on cultivated Schwann cells stressed by oxygen glucose deprivation (OGD) were studied. The negative effect of minocycline on Wallerian degeneration, the essential initial phase of degeneration/regeneration after nerve injury, that was recently demonstrated, was excluded by using predegenerated nerve and Schwann cell-enriched muscle grafts, both free of Wallerian degeneration. They were compared with common nerve grafts. The principle findings were that in vitro minocycline provided protective effects against OGD-induced death of Schwann cells by preventing permeability of the mitochondrial membrane. It suppressed the OGD-mediated induction of HIF-1alpha and BAX, and stabilized/induced BCL-2. Cytochrome c release and cleavage of procaspase-3 were diminished; release and translocation of AIF and cytotoxic cleavage of actin into fractin were stopped. In common nerve grafts, minocycline, besides its direct anti-ischemic effect, hampered revascularization by down-regulation of MMP9 and VEGF prolonging ischemia and impeding macrophage recruitment. In bioartificial nerve grafts that were free of Wallerian degeneration and revealed lower immunogenicity, minocycline aided the regeneration process. Here, the direct anti-ischemic effect of minocycline on Schwann cells, which are mandatory for successful peripheral nerve regeneration, dominated the systemic anti-angiogenic/pro-ischemic effects. In common nerve grafts, however, where Wallerian degeneration is a prerequisite, the anti-angiogenic and macrophage-depressing effect is an obstacle for regeneration.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Medical Neurobiology, University of Magdeburg, Leipziger Strasse 44, D-39120 Magdeburg, Germany.
| | | | | |
Collapse
|
43
|
Allmeling C, Jokuszies A, Reimers K, Kall S, Choi CY, Brandes G, Kasper C, Scheper T, Guggenheim M, Vogt PM. Spider silk fibres in artificial nerve constructs promote peripheral nerve regeneration. Cell Prolif 2008; 41:408-20. [PMID: 18384388 DOI: 10.1111/j.1365-2184.2008.00534.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE In our study, we describe the use of spider silk fibres as a new material in nerve tissue engineering, in a 20-mm sciatic nerve defect in rats. MATERIALS AND METHODS We compared isogenic nerve grafts to vein grafts with spider silk fibres, either alone or supplemented with Schwann cells, or Schwann cells and matrigel. Controls, consisting of veins and matrigel, were transplanted. After 6 months, regeneration was evaluated for clinical outcome, as well as for histological and morphometrical performance. RESULTS Nerve regeneration was achieved with isogenic nerve grafts as well as with all constructs, but not in the control group. Effective regeneration by isogenic nerve grafts and grafts containing spider silk was corroborated by diminished degeneration of the gastrocnemius muscle and by good histological evaluation results. Nerves stained for S-100 and neurofilament indicated existence of Schwann cells and axonal re-growth. Axons were aligned regularly and had a healthy appearance on ultrastructural examination. Interestingly, in contrast to recently published studies, we found that bridging an extensive gap by cell-free constructs based on vein and spider silk was highly effective in nerve regeneration. CONCLUSION We conclude that spider silk is a viable guiding material for Schwann cell migration and proliferation as well as for axonal re-growth in a long-distance model for peripheral nerve regeneration.
Collapse
Affiliation(s)
- C Allmeling
- Department of Plastic, Hand- and Reconstructive Surgery, Medical School Hannover, Hannover, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vein Grafts Used as Nerve Conduits for Obstetrical Brachial Plexus Palsy Reconstruction. Plast Reconstr Surg 2007; 120:1930-1941. [DOI: 10.1097/01.prs.0000287391.12943.00] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
45
|
Su Y, Zeng BF, Zhang CQ, Zhang KG, Xie XT. Study of biocompatibility of small intestinal submucosa (SIS) with Schwann cells in vitro. Brain Res 2007; 1145:41-7. [PMID: 17367764 DOI: 10.1016/j.brainres.2007.01.138] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2006] [Revised: 01/10/2007] [Accepted: 01/17/2007] [Indexed: 01/12/2023]
Abstract
No satisfactory method currently exists for repairing long peripheral nerve defects. Efforts have been made to fabricate bioactive artificial nerve conduits, comprised of a biomaterial pre-seeded with Schwann cells (SCs), which creating a favorable micro-environment for axonal regeneration, to be an alternative to autografting by means of tissue engineering. Small intestinal submucosa (SIS) possesses special biological characteristics and is comprehensively researched for tissue repairing at varied tissues and organs. This study investigated the biocompatibility of SIS with SCs in vitro. Cultured rat SCs were seeded on SIS. Cell morphology was observed by light microscopy, scanning electron microscopy and transmission electron microscope. The viability of SCs was measured by MTT assay. Secretion of NGF-beta and BDNF was quantitatively assessed by ELISA, and NGF-beta mRNA and BDNF mRNA were semi-quantitatively assessed by RT-PCR. The results indicated that SCs could adhere, migrate and proliferate on the surface of SIS in good condition with productive function of secreting growth factors. SIS has a good biocompatibility with SCs and SIS pre-seeded with SCs has potential to be an alternate candidate of autografting for repairing long peripheral nerve defects.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Biocompatible Materials/therapeutic use
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Cell Adhesion/physiology
- Cell Movement/physiology
- Cell Proliferation
- Cell Survival/physiology
- Cells, Cultured
- Graft Survival/physiology
- Intestinal Mucosa/physiology
- Intestinal Mucosa/ultrastructure
- Intestine, Small/physiology
- Intestine, Small/ultrastructure
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Nerve Growth Factor/genetics
- Nerve Growth Factor/metabolism
- Nerve Regeneration/physiology
- Peripheral Nerves/cytology
- Peripheral Nerves/physiology
- Peripheral Nerves/surgery
- Peripheral Nervous System Diseases/therapy
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Schwann Cells/metabolism
- Schwann Cells/ultrastructure
- Sus scrofa
- Tissue Engineering/methods
Collapse
Affiliation(s)
- Yan Su
- Department of Orthopaedics, The Sixth Affiliated People's Hospital, Shanghai Jiaotong University, 600 Yishan Road, Shanghai 200233, China
| | | | | | | | | |
Collapse
|
46
|
Hess JR, Brenner MJ, Fox IK, Nichols CM, Myckatyn TM, Hunter DA, Rickman SR, Mackinnon SE. Use of cold-preserved allografts seeded with autologous Schwann cells in the treatment of a long-gap peripheral nerve injury. Plast Reconstr Surg 2007; 119:246-259. [PMID: 17255680 DOI: 10.1097/01.prs.0000245341.71666.97] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Limitations in autogenous tissue have inspired the study of alternative materials for repair of complex peripheral nerve injuries. Cadaveric allografts are one potential reconstructive material, but their use requires systemic immunosuppression. Cold preservation (> or =7 weeks) renders allografts devoid of antigens, but these acellular substrates generally fail in supporting regeneration beyond 3 cm. In this study, the authors evaluated the reconstruction of extensive nonhuman primate peripheral nerve defects using 7-week cold-preserved allografts repopulated with cultured autologous Schwann cells. METHODS Ten outbred Macaca fascicularis primates were paired based on maximal genetic disparity as measured by similarity index assay. A total of 14 ulnar nerve defects measuring 6 cm were successfully reconstructed using autografts (n = 5), fresh allografts (n = 2), cold-preserved allografts (n = 3), or cold-preserved allografts seeded with autogenous Schwann cells (n = 4). Recipient immunoreactivity was evaluated by means of enzyme-linked immunosorbent spot assay, and nerves were harvested at 6 months for histologic and histomorphometric analysis. RESULTS Cytokine production in response to cold-preserved allografts and cold-preserved allografts seeded with autologous Schwann cells was similar to that observed for autografts. Schwann cell-repopulated cold-preserved grafts demonstrated significantly enhanced fiber counts, nerve density, and percentage nerve (p < 0.05) compared with unseeded cold-preserved grafts at 6 months after reconstruction. CONCLUSIONS Cold-preserved allografts seeded with autologous Schwann cells were well-tolerated in unrelated recipients and supported significant regeneration across 6-cm peripheral nerve defects. Use of cold-preserved allogeneic nerve tissue supplemented with autogenous Schwann cells poses a potentially safe and effective alternative to the use of autologous tissue in the reconstruction of extensive nerve injuries.
Collapse
Affiliation(s)
- Jason R Hess
- St. Louis, Mo. From the Division of Plastic and Reconstructive Surgery, Department of Surgery, and Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Allmeling C, Jokuszies A, Reimers K, Kall S, Vogt PM. Use of spider silk fibres as an innovative material in a biocompatible artificial nerve conduit. J Cell Mol Med 2007; 10:770-7. [PMID: 16989736 PMCID: PMC3933158 DOI: 10.1111/j.1582-4934.2006.tb00436.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Defects of peripheral nerves still represent a challenge for surgical nerve reconstruction. Recent studies concentrated on replacement by artificial nerve conduits from different synthetic or biological materials. In our study, we describe for the first time the use of spider silk fibres as a new material in nerve tissue engineering. Schwann cells (SC) were cultivated on spider silk fibres. Cells adhered quickly on the fibres compared to polydioxanone monofilaments (PDS). SC survival and proliferation was normal in Live/Dead assays. The silk fibres were ensheathed completely with cells. We developed composite nerve grafts of acellularized veins, spider silk fibres and SC diluted in matrigel. These artificial nerve grafts could be cultivated in vitro for one week. Histological analysis showed that the cells were vital and formed distinct columns along the silk fibres. In conclusion, our results show that artificial nerve grafts can be constructed successfully from spider silk, acellularized veins and SC mixed with matrigel.
Collapse
Affiliation(s)
- Christina Allmeling
- Department of Plastic, Hand- and Reconstructive Surgery, Medical School Hannover, Podbielskistr. 380, D-30659 Hannover, Germany.
| | | | | | | | | |
Collapse
|
48
|
Nie X, Zhang YJ, Tian WD, Jiang M, Dong R, Chen JW, Jin Y. Improvement of peripheral nerve regeneration by a tissue-engineered nerve filled with ectomesenchymal stem cells. Int J Oral Maxillofac Surg 2007; 36:32-8. [PMID: 17169530 DOI: 10.1016/j.ijom.2006.06.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 02/27/2006] [Accepted: 06/20/2006] [Indexed: 11/28/2022]
Abstract
Ectomesenchymal stem cells (EMSCs) originate from the cranial neural crest. They are a potential source of neuronal and Schwann cells (SCs) of the peripheral nervous system (PNS) during embryonic development. The third passage of EMSCs enzymatically isolated from the mandibular processes of Sprague-Dawley rats were cultured in forskolin and bovine pituitary extract for 6 days to generate functional Schwann cell phenotypes. Next, 10-mm defects in the sciatic nerves were bridged with an autograft, tissue-engineered nerve filled with differentiated cells in collagen, or a PLGA conduit alone in 18 rats, and the nerve defects of another four rats were left untreated. The regenerated nerves were evaluated by the sciatic functional index (SFI) monthly and by histological analysis 4 months after grafting. The recovery index of the sciatic nerve improved significantly in the autograft and tissue-engineered nerve groups, both of which were superior to the PLGA group. In animals transplanted with the EMSCs, there was greater regeneration than with conduit alone during the same period of implantation. These results show that when EMSCs are transplanted to a peripheral nerve defect they differentiate into supportive cells that contribute to the promotion of axonal regeneration.
Collapse
Affiliation(s)
- X Nie
- Department of Oral Histology and Pathology, College of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Soria JM, Martínez Ramos C, Bahamonde O, García Cruz DM, Salmerón Sánchez M, García Esparza MA, Casas C, Guzmán M, Navarro X, Gómez Ribelles JL, García Verdugo JM, Monleón Pradas M, Barcia JA. Influence of the substrate's hydrophilicity on thein vitro Schwann cells viability. J Biomed Mater Res A 2007; 83:463-70. [PMID: 17477391 DOI: 10.1002/jbm.a.31297] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A series of polymeric biomaterials including poly (methyl acrylate) (PMA), chitosan (CHT), poly(ethyl acrylate) (PEA), poly(hydroxyethyl acrylate) (PHEA), and a series of random copolymers containing ethyl acrylate and hydroxyethyl acrylate monomeric units were tested in vitro as culture substrates and compared for their impact on the proliferation and expansion of Schwann cells (SCs). Immunocytochemical staining assay and scanning electron microscopy techniques were applied to perform a quantitative analysis to determine the correct maintenance of the cultured glial cells on the different biomaterials. The results strongly suggest that cell attachment and proliferation is influenced by the substrate's surface chemistry, and that hydrophobic biomaterials based on PMA, PEA, and the copolymers PEA and PHEA in a narrow composition window are suitable substrates to promote cell attachment and proliferation of SCs in vitro.
Collapse
Affiliation(s)
- J M Soria
- Fundación Hospital General Universitario de Valencia, Avda. Tres Cruces s/n, 46014 Valencia, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Keilhoff G, Prell T, Langnaese K, Mawrin C, Simon M, Fansa H, Nicholas AP. Expression pattern of peptidylarginine deiminase in rat and human Schwann cells. Dev Neurobiol 2007; 68:101-14. [DOI: 10.1002/dneu.20578] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|