1
|
Zhang Z, Ma M. Strategies to enhance the ability of nerve guidance conduits to promote directional nerve growth. Biomed Eng Online 2024; 23:40. [PMID: 38582838 PMCID: PMC10998375 DOI: 10.1186/s12938-024-01233-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
Severely damaged peripheral nerves will regenerate incompletely due to lack of directionality in their regeneration, leading to loss of nerve function. To address this problem, various nerve guidance conduits (NGCs) have been developed to provide guidance for nerve repair. However, their clinical application is still limited, mainly because its effect in promoting nerve repair is not as good as autologous nerve transplantation. Therefore, it is necessary to enhance the ability of NGCs to promote directional nerve growth. Strategies include preparing various directional structures on NGCs to provide contact guidance, and loading various substances on them to provide electrical stimulation or neurotrophic factor concentration gradient to provide directional physical or biological signals.
Collapse
Affiliation(s)
- Ziyue Zhang
- South China University of Technology School of Medicine, Guangzhou, China.
| | - Muyuan Ma
- South China University of Technology School of Medicine, Guangzhou, China
| |
Collapse
|
2
|
Cao S, Yihao W, Qi T, Xiong A, Liu P, Chen Y, Zeng H, Yu F, Weng J. Combination of stem cells and nerve guide conduit for the treatment of peripheral nerve injury: A meta-analysis. Muscle Nerve 2024; 69:227-238. [PMID: 38063327 DOI: 10.1002/mus.28018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 01/18/2024]
Abstract
INTRODUCTION/AIMS Many small-sized, single-center preclinical studies have investigated the benefits of introducing stem cells into the interior of nerve conduit. The aims of this meta-analysis are to review and contrast the effects of various types of stem cells in in vivo models used to reconstruct peripheral nerve injuries (PNIs) and to assess the reliability and stability of the available evidence. METHODS A systematic search was conducted using Cochrane Library, Embase, PubMed, and Web of Science to identify studies conducted from January 1, 2000, to September 21, 2022, and investigate stem cell therapy in peripheral nerve reconstruction animal models. Studies that met the relevant criteria were deemed eligible for this meta-analysis. RESULTS Fifty-five preclinical studies with a total of 1234 animals were incorporated. Stem cells demonstrated a positive impact on peripheral nerve regeneration at different follow-up times in the forest plots of five outcome indicators: compound muscle action potential (CMAP) amplitude, latency, muscle mass ratio, nerve conduction velocity, and sciatic functional index (SFI). In most comparisons, stem cell groups showed substantial differences compared with the control groups. The superior performance of adipose-derived stem cells (ADSCs) in terms of SFI, CMAP amplitude, and latency (p < .001) was identified. DISCUSSION The findings consistently demonstrated a favorable outcome in the reconstruction process when utilizing different groups of stem cells, as opposed to control groups where stem cells were not employed.
Collapse
Affiliation(s)
- Siyang Cao
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Wei Yihao
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Ao Xiong
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Peng Liu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Yingqi Chen
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Fei Yu
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Jian Weng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China
| |
Collapse
|
3
|
Alakpa EV, Bahrd A, Wiklund K, Andersson M, Novikov LN, Ljungberg C, Kelk P. Bioprinted Schwann and Mesenchymal Stem Cell Co-Cultures for Enhanced Spatial Control of Neurite Outgrowth. Gels 2023; 9:gels9030172. [PMID: 36975621 PMCID: PMC10048219 DOI: 10.3390/gels9030172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Bioprinting nerve conduits supplemented with glial or stem cells is a promising approach to promote axonal regeneration in the injured nervous system. In this study, we examined the effects of different compositions of bioprinted fibrin hydrogels supplemented with Schwann cells and mesenchymal stem cells (MSCs) on cell viability, production of neurotrophic factors, and neurite outgrowth from adult sensory neurons. To reduce cell damage during bioprinting, we analyzed and optimized the shear stress magnitude and exposure time. The results demonstrated that fibrin hydrogel made from 9 mg/mL of fibrinogen and 50IE/mL of thrombin maintained the gel’s highest stability and cell viability. Gene transcription levels for neurotrophic factors were significantly higher in cultures containing Schwann cells. However, the amount of the secreted neurotrophic factors was similar in all co-cultures with the different ratios of Schwann cells and MSCs. By testing various co-culture combinations, we found that the number of Schwann cells can feasibly be reduced by half and still stimulate guided neurite outgrowth in a 3D-printed fibrin matrix. This study demonstrates that bioprinting can be used to develop nerve conduits with optimized cell compositions to guide axonal regeneration.
Collapse
Affiliation(s)
- Enateri V Alakpa
- Department of Integrative Medical Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Anton Bahrd
- Department of Physics, Umeå University, SE-901 87 Umeå, Sweden
| | - Krister Wiklund
- Department of Physics, Umeå University, SE-901 87 Umeå, Sweden
| | | | - Lev N Novikov
- Department of Integrative Medical Biology, Umeå University, SE-901 87 Umeå, Sweden
| | - Christina Ljungberg
- Department of Surgical and Perioperative Science, Section of Hand and Plastic Surgery, Umeå University, SE-901 87 Umeå, Sweden
| | - Peyman Kelk
- Department of Integrative Medical Biology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
4
|
Functional Gait Assessment Using Manual, Semi-Automated and Deep Learning Approaches Following Standardized Models of Peripheral Nerve Injury in Mice. Biomolecules 2022; 12:biom12101355. [PMID: 36291564 PMCID: PMC9599622 DOI: 10.3390/biom12101355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Objective: To develop a standardized model of stretch−crush sciatic nerve injury in mice, and to compare outcomes of crush and novel stretch−crush injuries using standard manual gait and sensory assays, and compare them to both semi-automated as well as deep-learning gait analysis methods. Methods: Initial studies in C57/Bl6 mice were used to develop crush and stretch−crush injury models followed by histologic analysis. In total, 12 eight-week-old 129S6/SvEvTac mice were used in a six-week behavioural study. Behavioral assessments using the von Frey monofilament test and gait analysis recorded on a DigiGait platform and analyzed through both Visual Gait Lab (VGL) deep learning and standardized sciatic functional index (SFI) measurements were evaluated weekly. At the termination of the study, neurophysiological nerve conduction velocities were recorded, calf muscle weight ratios measured and histological analyses performed. Results: Histological evidence confirmed more severe histomorphological injury in the stretch−crush injured group compared to the crush-only injured group at one week post-injury. Von Frey monofilament paw withdrawal was significant for both groups at week one compared to baseline (p < 0.05), but not between groups with return to baseline at week five. SFI showed hindered gait at week one and two for both groups, compared to baseline (p < 0.0001), with return to baseline at week five. Hind stance width (HSW) showed similar trends as von Frey monofilament test as well as SFI measurements, yet hind paw angle (HPA) peaked at week two. Nerve conduction velocity (NCV), measured six weeks post-injury, at the termination of the study, did not show any significant difference between the two groups; yet, calf muscle weight measurements were significantly different between the two, with the stretch−crush group demonstrating a lower (poorer) weight ratio relative to uninjured contralateral legs (p < 0.05). Conclusion: Stretch−crush injury achieved a more reproducible and constant injury compared to crush-only injuries, with at least a Sunderland grade 3 injury (perineurial interruption) in histological samples one week post-injury in the former. However, serial behavioral outcomes were comparable between the two crush groups, with similar kinetics of recovery by von Frey testing, SFI and certain VGL parameters, the latter reported for the first time in rodent peripheral nerve injury. Semi-automated and deep learning-based approaches for gait analysis are promising, but require further validation for evaluation in murine hind-limb nerve injuries.
Collapse
|
5
|
Weiss JB, Phillips CJ, Malin EW, Gorantla VS, Harding JW, Salgar SK. Stem cell, Granulocyte-Colony Stimulating Factor and/or Dihexa to promote limb function recovery in a rat sciatic nerve damage-repair model: Experimental animal studies. Ann Med Surg (Lond) 2021; 71:102917. [PMID: 34703584 PMCID: PMC8524106 DOI: 10.1016/j.amsu.2021.102917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Background Optimizing nerve regeneration and re-innervation of target muscle/s is the key for improved functional recovery following peripheral nerve damage. We investigated whether administration of mesenchymal stem cell (MSC), Granulocyte-Colony Stimulating Factor (G-CSF) and/or Dihexa can improve recovery of limb function following peripheral nerve damage in rat sciatic nerve transection-repair model. Materials and methods There were 10 experimental groups (n = 6–8 rats/group). Bone marrow derived syngeneic MSCs (2 × 106; passage≤6), G-CSF (200–400 μg/kg b.wt.), Dihexa (2–4 mg/kg b.wt.) and/or Vehicle were administered to male Lewis rats locally via hydrogel at the site of nerve repair, systemically (i.v./i.p), and/or to gastrocnemius muscle. The limb sensory and motor functions were assessed at 1–2 week intervals post nerve repair until the study endpoint (16 weeks). Results The sensory function in all nerve boundaries (peroneal, tibial, sural) returned to nearly normal by 8 weeks (Grade 2.7 on a scale of Grade 0–3 [0 = No function; 3 = Normal function]) in all groups combined. The peroneal nerve function recovered quickly with return of function at one week (∼2.0) while sural nerve function recovered rather slowly at four weeks (∼1.0). Motor function at 8–16 weeks post-nerve repair as determined by walking foot print grades significantly (P < 0.05) improved with MSC + G-CSF or MSC + Dihexa administrations into gastrocnemius muscle and mitigated foot flexion contractures. Conclusions These findings demonstrate MSC, G-CSF and Dihexa are promising candidates for adjunct therapies to promote limb functional recovery after surgical nerve repair, and have implications in peripheral nerve injury and limb transplantation. IACUC No.215064. G-CSF in combination with MSCs improved limb function recovery in sciatic nerve transection- repair model. Dihexa in combination with MSC improved limb function recovery in sciatic nerve transection- repair model. Foot flexion contractures were reduced with G-CSF & MSC or Dihexa & MSC administration into target muscle gastrocnemius. MSC, G-CSF or Dihexa combination therapy is attractive, feasible & promising in peripheral nerve injury repair and have implications in limb transplantation. The findings warrant further investigation to understand the cellular/molecular mechanisms.
Collapse
Affiliation(s)
- Jessica B Weiss
- Department of Surgery, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| | - Cody J Phillips
- Department of Surgery, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| | - Edward W Malin
- Department of Surgery, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| | - Vijay S Gorantla
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Joseph W Harding
- Department of Integrative Physiology & Neuroscience, Washington State University, Pullman, WA, USA
| | - Shashikumar K Salgar
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, Fort Lewis, Washington, USA
| |
Collapse
|
6
|
Zeng XX, Zeng J, Zhu B. Future generation of combined multimodal approach to treat brain glioblastoma multiforme and potential impact on micturition control. Rev Neurosci 2021; 33:313-326. [PMID: 34529907 DOI: 10.1515/revneuro-2021-0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/26/2021] [Indexed: 11/15/2022]
Abstract
Glioblastoma remains lethal even when treated with standard therapy. This review aims to outline the recent development of various advanced therapeutics for glioblastoma and briefly discuss the potential impact of glioblastoma and some of its therapeutic approaches on the neurological function micturition control. Although immunotherapy led to success in treating hematological malignancies, but no similar success occurred in treatment for brain glioblastoma. Neither regenerative medicine nor stem cell therapy led to astounding success in glioblastoma. However, CRISPR Cas system holds potential in multiple applications due to its capacity to knock-in and knock-out genes, modify immune cells and cell receptors, which will enable it to address clinical challenges in immunotherapy such as CAR-T and regenerative therapy for brain glioblastoma, improving the precision and safety of these approaches. The studies mentioned in this review could indicate that glioblastoma is a malignant disease with multiple sophisticated barriers to be overcome and more challenges might arise in the attempt of researchers to yield a successful cure. A multimodal approach of future generation of refined and safe therapeutics derived from CRISPR Cas therapeutics, immunotherapy, and regenerative therapeutics mentioned in this review might prolong survival or even contribute towards a potential cure for glioblastoma.
Collapse
Affiliation(s)
- Xiao Xue Zeng
- Guangzhou United Family Hospital, Fangyuan Road 28, Haizhu District, Guangzhou, Postcode: 510000, Guangdong Province, P. R. China
| | - Jianwen Zeng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Yinquan Road B24, Qingyuan City, Postcode: 511500, Guangdong Province, P. R. China
| | - Baoyi Zhu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Yinquan Road B24, Qingyuan City, Postcode: 511500, Guangdong Province, P. R. China
| |
Collapse
|
7
|
Ahmed MN, Shi D, Dailey MT, Rothermund K, Drewry MD, Calabrese TC, Cui XT, Syed-Picard FN. Dental Pulp Cell Sheets Enhance Facial Nerve Regeneration via Local Neurotrophic Factor Delivery. Tissue Eng Part A 2021; 27:1128-1139. [PMID: 33164704 PMCID: PMC8616747 DOI: 10.1089/ten.tea.2020.0265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An effective strategy for sustained neurotrophic factor (NTF) delivery to sites of peripheral nerve injury (PNI) would accelerate healing and enhance functional recovery, addressing the major clinical challenges associated with the current standard of care. In this study, scaffold-free cell sheets were generated using human dental pulp stem/progenitor cells, that endogenously express high levels of NTFs, for use as bioactive NTF delivery systems. Additionally, the effect of fibroblast growth factor 2 (FGF2) on NTF expression by dental pulp cell (DPC) sheets was evaluated. In vitro analysis confirmed that DPC sheets express high levels of NTF messenger RNA (mRNA) and proteins, and the addition of FGF2 to DPC sheet culture increased total NTF production by significantly increasing the cellularity of sheets. Furthermore, the DPC sheet secretome stimulated neurite formation and extension in cultured neuronal cells, and these functional effects were further enhanced when DPC sheets were cultured with FGF2. These neuritogenic results were reversed by NTF inhibition substantiating that DPC sheets have a positive effect on neuronal cell activity through the production of NTFs. Further evaluation of DPC sheets in a rat facial nerve crush injury model in vivo established that in comparison with untreated controls, nerves treated with DPC sheets had greater axon regeneration through the injury site and superior functional recovery as quantitatively assessed by compound muscle action potential measurements. This study demonstrates the use of DPC sheets as vehicles for NTF delivery that could augment the current methods for treating PNIs to accelerate regeneration and enhance the functional outcome. Impact statement The major challenges associated with current treatments of peripheral nerve injuries (PNIs) are prolonged repair times and insufficient functional recovery. Dental pulp stem/progenitor cells (DPCs) are known to endogenously express high levels of neurotrophic factors (NTFs), growth factors that enhance axon regeneration. In this study, we demonstrate that scaffold-free DPC sheets can act as effective carrier systems to facilitate the delivery and retention of NTF-producing DPCs to sites of PNIs and improve functional nerve regeneration. DPC sheets have high translational feasibility and could augment the current standard of care to enhance the quality of life for patients dealing with PNIs.
Collapse
Affiliation(s)
- Meer N. Ahmed
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Delin Shi
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew T. Dailey
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kristi Rothermund
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michelle D. Drewry
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tia C. Calabrese
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xinyan T. Cui
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fatima N. Syed-Picard
- Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania USA
- Address correspondence to: Fatima N. Syed-Picard, MSE, PhD, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, 413 Salk Pavilion, 355 Sutherland Drive, Pittsburgh, PA 15213, USA
| |
Collapse
|
8
|
Moore JT, Wier CG, Lemmerman LR, Ortega-Pineda L, Dodd DJ, Lawrence WR, Duarte-Sanmiguel S, Dathathreya K, Diaz-Starokozheva L, Harris HN, Sen CK, Valerio IL, Higuita-Castro N, Arnold WD, Kolb SJ, Gallego-Perez D. Nanochannel-Based Poration Drives Benign and Effective Nonviral Gene Delivery to Peripheral Nerve Tissue. ADVANCED BIOSYSTEMS 2020; 4:e2000157. [PMID: 32939985 PMCID: PMC7704786 DOI: 10.1002/adbi.202000157] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/04/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023]
Abstract
While gene and cell therapies have emerged as promising treatment strategies for various neurological conditions, heavy reliance on viral vectors can hamper widespread clinical implementation. Here, the use of tissue nanotransfection as a platform nanotechnology to drive nonviral gene delivery to nerve tissue via nanochannels, in an effective, controlled, and benign manner is explored. TNT facilitates plasmid DNA delivery to the sciatic nerve of mice in a voltage-dependent manner. Compared to standard bulk electroporation (BEP), impairment in toe-spread and pinprick response is not caused by TNT, and has limited to no impact on electrophysiological parameters. BEP, however, induces significant nerve damage and increases macrophage immunoreactivity. TNT is subsequently used to deliver vasculogenic cell therapies to crushed nerves via delivery of reprogramming factor genes Etv2, Foxc2, and Fli1 (EFF). The results indicate the TNT-based delivery of EFF in a sciatic nerve crush model leads to increased vascularity, reduced macrophage infiltration, and improved recovery in electrophysiological parameters compared to crushed nerves that are TNT-treated with sham/empty plasmids. Altogether, the results indicate that TNT can be a powerful platform nanotechnology for localized nonviral gene delivery to nerve tissue, in vivo, and the deployment of reprogramming-based cell therapies for nerve repair/regeneration.
Collapse
Affiliation(s)
- Jordan T. Moore
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Luke R. Lemmerman
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Daniel J. Dodd
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - William R. Lawrence
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Kavya Dathathreya
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | | | - Hallie N. Harris
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chandan K. Sen
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Ian L. Valerio
- Plastic and Reconstructive Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - W. David Arnold
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Stephen J. Kolb
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
9
|
|
10
|
Kubiak CA, Grochmal J, Kung TA, Cederna PS, Midha R, Kemp SWP. Stem-cell-based therapies to enhance peripheral nerve regeneration. Muscle Nerve 2019; 61:449-459. [PMID: 31725911 DOI: 10.1002/mus.26760] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022]
Abstract
Peripheral nerve injury remains a major cause of morbidity in trauma patients. Despite advances in microsurgical techniques and improved understanding of nerve regeneration, obtaining satisfactory outcomes after peripheral nerve injury remains a difficult clinical problem. There is a growing body of evidence in preclinical animal studies demonstrating the supportive role of stem cells in peripheral nerve regeneration after injury. The characteristics of both mesoderm-derived and ectoderm-derived stem cell types and their role in peripheral nerve regeneration are discussed, specifically focusing on the presentation of both foundational laboratory studies and translational applications. The current state of clinical translation is presented, with an emphasis on both ethical considerations of using stems cells in humans and current governmental regulatory policies. Current advancements in cell-based therapies represent a promising future with regard to supporting nerve regeneration and achieving significant functional recovery after debilitating nerve injuries.
Collapse
Affiliation(s)
- Carrie A Kubiak
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan
| | - Joey Grochmal
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Theodore A Kung
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan
| | - Paul S Cederna
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Rajiv Midha
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stephen W P Kemp
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
11
|
Modrak M, Talukder MAH, Gurgenashvili K, Noble M, Elfar JC. Peripheral nerve injury and myelination: Potential therapeutic strategies. J Neurosci Res 2019; 98:780-795. [PMID: 31608497 DOI: 10.1002/jnr.24538] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/30/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022]
Abstract
Traumatic peripheral nerve injury represents a major clinical and public health problem that often leads to significant functional impairment and permanent disability. Despite modern diagnostic procedures and advanced microsurgical techniques, functional recovery after peripheral nerve repair is often unsatisfactory. Therefore, there is an unmet need for new therapeutic or adjunctive strategies to promote the functional recovery in nerve injury patients. In contrast to the central nervous system, Schwann cells in the peripheral nervous system play a pivotal role in several aspects of nerve repair such as degeneration, remyelination, and axonal growth. Several non-surgical approaches, including pharmacological, electrical, cell-based, and laser therapies, have been employed to promote myelination and enhance functional recovery after peripheral nerve injury. This review will succinctly discuss the potential therapeutic strategies in the context of myelination following peripheral neurotrauma.
Collapse
Affiliation(s)
- Max Modrak
- School of Medicine & Dentistry, The University of Rochester Medical Center, Rochester, New York, USA
| | - M A Hassan Talukder
- Department of Orthopaedics & Rehabilitation, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Khatuna Gurgenashvili
- Department of Neurology, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mark Noble
- Department of Biomedical Genetics, The University of Rochester Medical Center, Rochester, New York, USA
| | - John C Elfar
- Department of Orthopaedics & Rehabilitation, Penn State Hershey College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
12
|
Saez DM, Sasaki RT, Martins DDO, Chacur M, Kerkis I, da Silva MCP. Rat Facial Nerve Regeneration with Human Immature Dental Pulp Stem Cells. Cell Transplant 2019; 28:1573-1584. [PMID: 31462071 PMCID: PMC6923557 DOI: 10.1177/0963689719854446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Facial paralysis can result in severe implications for the patients. However, stem cell
biology has become an important field in regenerative medicine since the discovery and
characterization of mesenchymal stem cells. Our aim was to evaluate the regeneration after
facial nerve crush injury and application of human immature dental pulp stem cells
(iDPSC). For this study 70 Wistar rats underwent a unilateral facial nerve crush injury
and were divided into two groups: Group I (GI): Crushed; Group II (GII): Crushed and
iDPSC, and distributed into study periods of 3, 7, 14, 21, and 42 postoperative days.
Facial nerve regeneration was analyzed via functional recovery of whisker movement,
histomorphometric analysis, and immunoblotting assay. The results show that GII had
complete functional recovery at 14 days, while GI recovered after 42 days. Also, regarding
the facial nerve trunk, GII presented histological improvement, evidencing better axonal
and structural organization of the myelin sheath, and exhibited statistically higher
values for the outer and inner perimeters and g-ratio. Nevertheless, GI exhibited
statistically higher values for the thickness of myelin sheath. In the buccal branch, no
differences were observed for all parameters between groups. At 42 days, both groups GI
and GII were close to the levels observed for the control group. Concerning nerve growth
factor expression, GII exhibited statistically greater values (p <
0.05) compared with the control group at 7 days. In summary, a single injection of human
iDPSC promoted a positive effect on regeneration of the facial nerve trunk after 14 days
and provided an alternative to support regeneration following peripheral nerve injury.
Collapse
Affiliation(s)
- Daniel Martinez Saez
- Department of Morphology and Genetics, Universidade Federal de São Paulo,
São Paulo, Brazil
- Daniel Martinez Saez, Department of Morphology and
Genetics, Universidade Federal de São Paulo, Rua Botucatu, 740 - Edifício
Leitão da Cunha, Vila Clementino, São Paulo 04023, Brazil.
| | - Robson Tetsuo Sasaki
- Department of Morphology and Genetics, Universidade Federal de São Paulo,
São Paulo, Brazil
| | | | - Marucia Chacur
- Departament of Anatomy, Institute of Biomedical Sciences – Universidade de
São Paulo, São Paulo, Brazil
| | - Irina Kerkis
- Department of Genetics, Instituto Butantan, São Paulo, Brazil
| | | |
Collapse
|
13
|
Bingham JR, Kniery KR, Jorstad NL, Horkayne-Szakaly I, Hoffer ZS, Salgar SK. "Stem cell therapy to promote limb function recovery in peripheral nerve damage in a rat model" - Experimental research. Ann Med Surg (Lond) 2019; 41:20-28. [PMID: 31011420 PMCID: PMC6463551 DOI: 10.1016/j.amsu.2019.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/26/2019] [Accepted: 03/24/2019] [Indexed: 02/06/2023] Open
Abstract
Background Optimizing nerve regeneration and mitigating muscle atrophy are the keys to successful outcomes in peripheral nerve damage. We investigated whether mesenchymal stem cell (MSC) therapy can improve limb function recovery in peripheral nerve damage. Materials and methods We used sciatic nerve transection/repair (SNR) and individual nerve transection/repair (INR; branches of sciatic nerve - tibial, peroneal, sural) models to study the effect of MSCs on proximal and distal peripheral nerve damages, respectively, in male Lewis rats. Syngeneic MSCs (5 × 106; passage≤6) or saline were administered locally and intravenously. Sensory/motor functions (SF/MF) of the limb were assessed. Results Rat MSCs (>90%) were CD29+, CD90+, CD34−, CD31− and multipotent. Total SF at two weeks post-SNR & INR with or without MSC therapy was ∼1.2 on a 0–3 grading scale (0 = No function; 3 = Normal); by 12 weeks it was 2.6–2.8 in all groups (n ≥ 9/group). MSCs accelerated SF onset. At eight weeks post-INR, sciatic function index (SFI), a measure of MF (0 = Normal; −100 = Nonfunctional) was −34 and −77 in MSC and vehicle groups, respectively (n ≥ 9); post-SNR it was −72 and −92 in MSC and vehicle groups, respectively. Long-term MF (24 weeks) was apparent in MSC treated INR (SFI -63) but not in SNR (SFI -100). Gastrocnemius muscle atrophy was significantly reduced (P < 0.05) in INR. Nerve histomorphometry revealed reduced axonal area (P < 0.01) but no difference in myelination (P > 0.05) in MSC treated INR compared to the naive contralateral nerve. Conclusion MSC therapy in peripheral nerve damage appears to improve nerve regeneration, mitigate flexion-contractures, and promote limb functional recovery. Mesenchymal stem cell (MSC) therapy improved limb functional recovery. MSCs improved nerve regeneration and mitigated foot flexion-contractures. Limb muscle atrophy was significantly reduced in individual nerve repair (INR). Functional recovery in distal nerve repair (INR) was superior to proximal (SNR). MSC therapy is attractive, feasible & promising in peripheral nerve injury repair.
Collapse
Affiliation(s)
- Jason R Bingham
- Department of Surgery, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| | - Kevin R Kniery
- Department of Surgery, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| | - Nikolas L Jorstad
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Iren Horkayne-Szakaly
- Department of Neuropathology & Ophthalmic Pathology, Joint Pathology Center, Defense Health Agency, Silver Spring, MD, 20910, USA
| | - Zachary S Hoffer
- Department of Pathology, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| | - Shashikumar K Salgar
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, 98431, USA
| |
Collapse
|
14
|
Ge Y, Zhang Y, Tang Q, Gao J, Yang H, Gao Z, Zhao RC. Mechanisms of the Immunomodulation Effects of Bone Marrow-Derived Mesenchymal Stem Cells on Facial Nerve Injury in Sprague-Dawley Rats. Stem Cells Dev 2019; 28:489-496. [PMID: 30704338 DOI: 10.1089/scd.2018.0104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Normal facial nerve (FN) function is very important for human being. However, if injured, FN function is difficult to restore completely. Recently, many studies reported the immune regulation function of stem cells (SCs). However, the immunomodulation function of SCs on FN injury is still unclear. Our study aims to explore the mechanism of immunomodulation effect of Sprague-Dawley rat bone marrow-derived SCs (BMSCs) on FN injury and specially focus on the regulation of Th17 and the protection effects of BMSCs on central facial motor neurons (FMNs). First, rat FNs were harvested. FN and BMSCs were cultured together or separately and levels of transforming growth factor (TGF)-β1, interleukin (IL)-6, hepatocyte growth factor (HGF), inducible nitric oxide synthase (iNOS), and prostaglandin E2 (PGE2) in supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Then, after treating with or without local BMSCs injection, the proportion of Th17 in neck lymph nodes (LNs) was investigated in rat FN injury models. Furthermore, the apoptotic index of FMNs was studied in rat FN injury models that were treated with or without BMSCs. We found that BMSCs could secrete high levels of IL-6, HGF, PGE2, iNOS, and TGF-β1 in culture. The percentage of Th17 of neck LNs in BMSCs-treated group was significantly lower than that in the control group. The apoptotic index of FMNs in BMSCs-treated group was significantly lower than that in the control group. In conclusion, our research indicates BMSCs could independently secrete cytokines IL-6, HGF, PGE2, iNOS, and TGF-β1, and these cytokines could regulate the balance among subsets of CD4+ T cells and could protect FMNs by inhibiting neuron apoptosis.
Collapse
Affiliation(s)
- Yining Ge
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,3 Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongli Zhang
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Tang
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juanjuan Gao
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Yang
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiqiang Gao
- 1 Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,2 Department of Otolaryngology, Translational Medicine Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Robert Chunhua Zhao
- 4 Department of Cell Biology, Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Bucan V, Fliess M, Schnabel R, Peck CT, Vaslaitis D, Fülbier A, Reimers K, Strauss S, Vogt PM, Radtke C. In vitro enhancement and functional characterization of neurite outgrowth by undifferentiated adipose-derived stem cells. Int J Mol Med 2018; 43:593-602. [PMID: 30431135 DOI: 10.3892/ijmm.2018.3979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/22/2018] [Indexed: 11/05/2022] Open
Abstract
Adipose‑derived stem cells (ASCs) can easily be obtained and expanded in vitro for use in autologous cell therapy. Via their production of cytokines and neurotrophic factors, transplanted ASCs provide neuroprotection, neovascularization and induction of axonal sprouting. However, the influencing mechanism of undifferentiated ASCs on nerve regeneration is currently only partially understood. In the present study, undifferentiated ASCs and cutaneous primary afferent dorsal root ganglion (DRG) neurons were co‑cultured in order to investigate their interaction. ASCs were isolated from adult rat fat tissue. The presence of characteristic stem cell markers was determined by flow cytometry in three subsequent passages. Adipogenic, osteogenic, chondrogenic and glial differentiation was performed in order to evaluate their differentiation capacity. A direct co‑culture system with DRG cells was established to determine the effect of undifferentiated pluripotent ASCs on neurite elongation. Neurite outgrowth, length and number was examined in the co‑culture and compared with single‑culture cells and cells stimulated with nerve growth factor (NGF). In ASC cultures, NGF expression was assessed by ELISA. The present results demonstrated that the specific mesenchymal stem cell surface markers CD44, CD73 and CD90 were detected in all three subsequent passages of the isolated ASCs. In accordance, ASC differentiation into adipogenic, osteogenic, chondrogenic and Schwann cell phenotype was conducted successfully. Neurite outgrowth of DRG neurons was enhanced following co‑culture with ASCs, resulting in increased neurite length after 24 h of cultivation. Furthermore, neurite outgrowth of DRG neurons was directed towards the undifferentiated ASC and direct cell‑to‑cell contact was observed. In summary, the results of the present study revealed an interaction between the two cell types with guidance of neurite growth towards the undifferentiated ASC. These findings suggest that the use of undifferentiated ASC optimizing tissue‑engineered constructs may be promising for peripheral nerve repair.
Collapse
Affiliation(s)
- Vesna Bucan
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Malte Fliess
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Reinhild Schnabel
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Claas-Tido Peck
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Desiree Vaslaitis
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Angela Fülbier
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Kerstin Reimers
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Sarah Strauss
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Peter M Vogt
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| | - Christine Radtke
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, D‑30625 Hannover, Germany
| |
Collapse
|
16
|
Petrova ES. Differentiation Potential of Mesenchymal Stem Cells and Stimulation of Nerve Regeneration. Russ J Dev Biol 2018. [DOI: 10.1134/s1062360418040033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
17
|
Petrova ES, Isaeva EN, Kolos EA, Korzhevskii DE. Vascularization of the Damaged Nerve under the Effect of Experimental Cell Therapy. Bull Exp Biol Med 2018; 165:161-165. [PMID: 29797137 DOI: 10.1007/s10517-018-4120-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Indexed: 10/16/2022]
Abstract
Quantitative analysis of blood vessels in the distal segment of rat sciatic nerve after its ligation for 40 sec and subperineurial administration of mesenchymal stem cells or dissociated cells of rat embryonic spinal cord was carried our by immunohistochemical tracing of von Willebrand factor, a marker of endothelial cells of blood vessels. It was found that the number of blood vessels per unit area of the nerve trunk in 21 days after injury and administration of mesenchymal stem cells increased by more than 1.5 times in comparison with the control (damaged nerve). After administration of dissociated cells of the embryonic spinal cord, this effect was not observed. It is assumed that mesenchymal stem cells stimulate the growth of vessels of the damaged nerve via production of angiogenic factors.
Collapse
Affiliation(s)
- E S Petrova
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Research Institute of Experimental Medicine, St. Petersburg, Russia.
| | - E N Isaeva
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Research Institute of Experimental Medicine, St. Petersburg, Russia
| | - E A Kolos
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Research Institute of Experimental Medicine, St. Petersburg, Russia
| | - D E Korzhevskii
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Research Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
18
|
Aligned contiguous microfiber platform enhances neural differentiation of embryonic stem cells. Sci Rep 2018; 8:6087. [PMID: 29666444 PMCID: PMC5904125 DOI: 10.1038/s41598-018-24522-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/03/2018] [Indexed: 11/09/2022] Open
Abstract
A microfiber platform that is able to enhance neuronal differentiation and guide aligned neurite outgrowths is essential to the repair of nerve damage. To achieve this aim, we utilized biocompatible and biodegradable poly lactic-co-glycolic acid (PLGA) to design a novel Aligned Contiguous Microfiber Platform (ACMFP) as substrates for the neuronal induction of mouse embryonic stem (ES) cells. To generate the ACMFP, a modified micro-fluid chip system was established to control microfiber parameters including fiber diameter, alignment, and the distance between fibers. Further, Pluronic-F127 was applied to the ACMFP system to maintain a stable and highly aligned fiber platform for at least 12 days. We found that the ACMFP can enhance the neuronal differentiation of mouse ES cells. The ACMFP system showed significantly better neurite outgrowth alignment guidance compared to the control substrate. The effects of alignment guidance were inversely proportionate to the diameter of the fiber, with the optimal diameter size of 60 µm. This study demonstrates a novel ACMFP system that can be used as a biomaterial substrate for neurite outgrowth alignment guidance, which may provide a new model for the development of a multidisciplinary treatment option for nerve injuries.
Collapse
|
19
|
Cui Y, Yao Y, Zhao Y, Xiao Z, Cao Z, Han S, Li X, Huan Y, Pan J, Dai J. Functional collagen conduits combined with human mesenchymal stem cells promote regeneration after sciatic nerve transection in dogs. J Tissue Eng Regen Med 2018; 12:1285-1296. [PMID: 29499096 DOI: 10.1002/term.2660] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 01/08/2018] [Accepted: 02/17/2018] [Indexed: 12/20/2022]
Abstract
Numerous studies have focused on the development of novel and innovative approaches for the treatment of peripheral nerve injury using artificial nerve guide conduits. In this study, we attempted to bridge 3.5-cm defects of the sciatic nerve with a longitudinally oriented collagen conduit (LOCC) loaded with human umbilical cord mesenchymal stem cells (hUC-MSCs). The LOCC contains a bundle of longitudinally aligned collagenous fibres enclosed in a hollow collagen tube. Our previous studies showed that an LOCC combined with neurotrophic factors enhances peripheral nerve regeneration. However, it remained unknown whether an LOCC seeded with hUC-MSCs could also promote regeneration. In this study, using various histological and electrophysiological analyses, we found that an LOCC provides mechanical support to newly growing nerves and functions as a structural scaffold for cells, thereby stimulating sciatic nerve regeneration. The LOCC and hUC-MSCs synergistically promoted regeneration and improved the functional recovery in a dog model of sciatic nerve injury. Therefore, the combined use of an LOCC and hUC-MSCs might have therapeutic potential for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Yi Cui
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, China.,Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yao Yao
- Department of Prosthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yannan Zhao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhifeng Xiao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zongfu Cao
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, China
| | - Sufang Han
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xing Li
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yong Huan
- State Key Laboratory of Nonlinear Mechanics (LNM), Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Juli Pan
- Department of Prosthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Jianwu Dai
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
20
|
Mirfeizi L, Stratton JA, Kumar R, Shah P, Agabalyan N, Stykel MG, Midha R, Biernaskie J, Kallos MS. Serum-free bioprocessing of adult human and rodent skin-derived Schwann cells: implications for cell therapy in nervous system injury. J Tissue Eng Regen Med 2017; 11:3385-3397. [PMID: 28176458 DOI: 10.1002/term.2252] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/19/2016] [Accepted: 07/03/2016] [Indexed: 02/06/2023]
Abstract
Peripheral nerve injury affects 2.8% of trauma patients with severe cases often resulting in long-lived permanent disability, despite nerve repair surgery. Autologous Schwann cell (SC) therapy currently provides an exciting avenue for improved outcomes for these patients, particularly with the possibility to derive SCs from easily-accessible adult skin. However, due to current challenges regarding the efficient expansion of these cells, further optimization is required before they can be seriously considered for clinical application. Here, a microcarrier-based bioreactor system is proposed as a means to scale-up large numbers of adult skin-derived SCs for transplantation into the injured nerve. Bioprocessing parameters that allow for the expansion of adult rodent SCs have been identified, whilst maintaining similar rates of proliferation (as compared to static-grown SCs), expression of SC markers, and, importantly, their capacity to myelinate axons following transplant into the injured sciatic nerve. The same bioprocessing parameters can be applied to SCs derived from adult human skin, and like rodent cells, they sustain their proliferative potential and expression of SC markers. Taken together, this dataset demonstrates the basis for a scalable bioprocess for the production of SCs, an important step towards clinical use of these cells as an adjunct therapy for nerve repair. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Leila Mirfeizi
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jo Anne Stratton
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ranjan Kumar
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Prajay Shah
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Natacha Agabalyan
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Morgan G Stykel
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
21
|
Fitzpatrick EB, Dehart MJ, Brown TA, Salgar SK. Mesenchymal stem cell therapy to promote limb transplant functional recovery. Microsurgery 2016; 37:222-234. [DOI: 10.1002/micr.30068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/31/2016] [Accepted: 04/22/2016] [Indexed: 12/13/2022]
Affiliation(s)
| | - Mary J. Dehart
- Department of Clinical Investigation; Madigan Army Medical Center; Tacoma WA 98431
| | - Tommy A. Brown
- Department of Surgery; Madigan Army Medical Center; Tacoma WA 98431
| | | |
Collapse
|
22
|
Petrova ES. Neurons with different neurotransmitters in embryonic neocortical allografts in the rat sciatic nerve. BIOL BULL+ 2016. [DOI: 10.1134/s1062359016020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Petrova ES, Kolos EA, Korzhevskii DE. Differentiation of Cholinergic Neurons in Rat Spinal Cord Under Conditions of Allotransplantation into a Peripheral Nerve and In Situ Development. Bull Exp Biol Med 2015; 160:141-7. [PMID: 26593413 DOI: 10.1007/s10517-015-3115-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Indexed: 12/23/2022]
Abstract
The method of ectopic transplantation of embryonic anlages of CNS allows studying histoblastic potencies of progenitor cells developing under conditions of changed microenvironment. Some progenitor cells in the transplants of rat embryonic spinal cord retained their ability to express choline acetyltransferase after transplantation into the sciatic nerve of adult animals. Comparative analysis of cholinergic neurons in the neurotransplants and neurons formed in rat spinal cord during normal ontogeny showed that choline acetyltransferase-positive cells after transplantation into the nerve reached morphological differentiation of motor neurons at later terms than cells developing in situ. They were scattered one by one and did not form nuclear nerve centers. We did not fi nd structures similar to presynaptic cholinergic buds typical of intact spinal cord near these cells throughout the observation period. Solitary cholinergic neurons survived in the transplants for 19 months.
Collapse
Affiliation(s)
- E S Petrova
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Institute of Experimental Medicine, St. Petersburg, Russia.
| | - E A Kolos
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Institute of Experimental Medicine, St. Petersburg, Russia
| | - D E Korzhevskii
- Laboratory of Functional Morphology of the Central and Peripheral Nervous System, Department of General and Special Morphology, Institute of Experimental Medicine, St. Petersburg, Russia
| |
Collapse
|
24
|
Fryer M, Grahammer J, Khalifian S, Furtmüller GJ, Lee WPA, Raimondi G, Brandacher G. Exploring cell-based tolerance strategies for hand and face transplantation. Expert Rev Clin Immunol 2015; 11:1189-204. [DOI: 10.1586/1744666x.2015.1078729] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
25
|
Grochmal J, Midha R. Recent advances in stem cell-mediated peripheral nerve repair. Cells Tissues Organs 2015; 200:13-22. [PMID: 25825283 DOI: 10.1159/000369450] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2014] [Indexed: 11/19/2022] Open
Abstract
A major advance in the field of peripheral nerve repair has been the advent of stem and progenitor cell use to supplement the regenerative environment in animal models of nerve injury. As Schwann cell replacements, stem cells may be even better suited to promoting regeneration in these scenarios. We review the recent literature detailing the search for the definitive Schwann cell replacement cell, including a look at genetic modification of transplanted cells for nerve injury repair.
Collapse
|
26
|
Petrova ES, Isaeva EN, Korzhevskii DE. Effect of allotransplants containing dissociated cells of rat embryonic spinal cord on nerve fiber regeneration in a recipient. Bull Exp Biol Med 2014; 158:123-6. [PMID: 25403413 DOI: 10.1007/s10517-014-2707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Indexed: 11/29/2022]
Abstract
Regeneration of nerve fibers in rat sciatic nerve was quantitatively assessed after injury (ligation) and injection of dissociated cells derived from embryonic spinal cord. A suspension of dissociated spinal cord cells from rat embryos was transplanted under the perineurium of a nerve trunk. After transplantation, bromodeoxyuridine-labeled precursor cells survived and retained the label for more than 2 months; some of these cells differentiated into NeuNpositive neurons. Analysis of semithin sections of the distal nerve segment from the recipient taken at a distance of 0.5 cm from the site of injury showed that transplantation of dissociated cells of embryonic spinal cord led to an increase in the number of myelinated nerve fibers in the recipient nerve.
Collapse
Affiliation(s)
- E S Petrova
- Laboratory of Functional Morphology of Central and Peripheral Nervous System, Department of General and Special Morphology, Research Institute of Experimental Medicine, North-Western Division of the Russian Academy of Medical Sciences, St. Petersburg, Russia,
| | | | | |
Collapse
|
27
|
Petrova ES, Isaeva EN. Study of effect of embryonic anlage allografts of the rat spinal cord on growth of regenerating fibers of the recipient nerve. BIOL BULL+ 2014. [DOI: 10.1134/s1062359014060089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Georgiou M, Golding JP, Loughlin AJ, Kingham PJ, Phillips JB. Engineered neural tissue with aligned, differentiated adipose-derived stem cells promotes peripheral nerve regeneration across a critical sized defect in rat sciatic nerve. Biomaterials 2014; 37:242-51. [PMID: 25453954 DOI: 10.1016/j.biomaterials.2014.10.009] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/02/2014] [Indexed: 01/12/2023]
Abstract
Adipose-derived stem cells were isolated from rats and differentiated to a Schwann cell-like phenotype in vitro. The differentiated cells (dADSCs) underwent self-alignment in a tethered type-1 collagen gel, followed by stabilisation to generate engineered neural tissue (EngNT-dADSC). The pro-regenerative phenotype of dADSCs was enhanced by this process, and the columns of aligned dADSCs in the aligned collagen matrix supported and guided neurite extension in vitro. EngNT-dADSC sheets were rolled to form peripheral nerve repair constructs that were implanted within NeuraWrap conduits to bridge a 15 mm gap in rat sciatic nerve. After 8 weeks regeneration was assessed using immunofluorescence imaging and transmission electron microscopy and compared to empty conduit and nerve graft controls. The proportion of axons detected in the distal stump was 3.5 fold greater in constructs containing EngNT-dADSC than empty tube controls. Our novel combination of technologies that can organise autologous therapeutic cells within an artificial tissue construct provides a promising new cellular biomaterial for peripheral nerve repair.
Collapse
Affiliation(s)
- Melanie Georgiou
- Advanced Centre for Biochemical Engineering, Bernard Katz Building, University College London, Gordon Street, London WC1H 0AH, UK; Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Jon P Golding
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Alison J Loughlin
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Paul J Kingham
- Department of Integrative Medical Biology, Umeå University, SE 901 87 Umeå, Sweden
| | - James B Phillips
- Department of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK; Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, 256 Gray's Inn Road, London WC1X 8LD, UK.
| |
Collapse
|
29
|
Petrova ES, Isaeva EN, Korzhevskii DE. Development of rat embryonic spinal ganglion cells in damaged nerve. Bull Exp Biol Med 2014; 157:637-40. [PMID: 25257430 DOI: 10.1007/s10517-014-2633-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Indexed: 10/24/2022]
Abstract
The development of dissociated cells from rat embryonic spinal ganglion after transplantation to damaged nerve of adult animals was studied using immunohistochemical differentiation markers of neural and glial cells. The cell suspension obtained after dissociation of rat embryonic spinal ganglia (embryonic day 15) was injected into the proximal segment of crushed sciatic nerve. The nerve was damaged by ligation for 40 sec. Progenitor cells were labeled with 5-bromo-2'-deoxyuridine (BrdU) before transplantation. BrdU-immunopositive cells were detected in the nerve trunks of recipients on days 1, 21, and 28 after transplantation. Dissociated cells of rat embryonic spinal ganglion (embryonic day 15) survived for at least 4 weeks after transplantation to the nerve and differentiate into NeuN-immunopositive neurons with morphological properties of sensory neurons and satellite cells containing S100 protein.
Collapse
Affiliation(s)
- E S Petrova
- Laboratory of Functional Morphology of Central and Peripheral Nervous System, Department of General and Special Morphology, Research Institute of Experimental Medicine, North-West Division of the Russian Academy of Medical Sciences, St. Petersburg, Russia,
| | | | | |
Collapse
|
30
|
Shakhbazau A, Mohanty C, Kumar R, Midha R. Sensory recovery after cell therapy in peripheral nerve repair: effects of naïve and skin precursor-derived Schwann cells. J Neurosurg 2014; 121:423-31. [DOI: 10.3171/2014.5.jns132132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Object
Cell therapy is a promising candidate among biological or technological innovations sought to augment microsurgical techniques in peripheral nerve repair. This report describes long-term functional regenerative effects of cell therapy in the rat injury model with a focus on sensory recovery.
Methods
Schwann cells were derived from isogenic nerve or skin precursor cells and injected into the transected and immediately repaired sciatic nerve distal to the injury site. Sensory recovery was assessed at weeks 4, 7, and 10. Axonal regeneration was assessed at Week 11.
Results
By Week 10, thermal sensitivity in cell therapy groups returned to a level indistinguishable from the baseline (p > 0.05). Immunohistochemistry at 11 weeks after injury showed improved regeneration of NF+ and IB4+ axons.
Conclusions:
The results of this study show that cell therapy significantly improves thermal sensation and the number of regenerated sensory neurons at 11 weeks after injury. These findings contribute to the view of skin-derived stem cells as a reliable source of Schwann cells with therapeutic potential for functional recovery in damaged peripheral nerve.
Collapse
Affiliation(s)
- Antos Shakhbazau
- 1Department of Clinical Neuroscience, Faculty of Medicine,
- 2Hotchkiss Brain Institute, and
| | | | - Ranjan Kumar
- 1Department of Clinical Neuroscience, Faculty of Medicine,
- 2Hotchkiss Brain Institute, and
- 3Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rajiv Midha
- 1Department of Clinical Neuroscience, Faculty of Medicine,
- 2Hotchkiss Brain Institute, and
| |
Collapse
|
31
|
Ribeiro J, Gartner A, Pereira T, Gomes R, Lopes MA, Gonçalves C, Varejão A, Luís AL, Maurício AC. Perspectives of employing mesenchymal stem cells from the Wharton's jelly of the umbilical cord for peripheral nerve repair. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 108:79-120. [PMID: 24083432 DOI: 10.1016/b978-0-12-410499-0.00004-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cells (MSCs) from Wharton's jelly present high plasticity and low immunogenicity, turning them into a desirable form of cell therapy for the injured nervous system. Their isolation, expansion, and characterization have been performed from cryopreserved umbilical cord tissue. Great concern has been dedicated to the collection, preservation, and transport protocols of the umbilical cord after the parturition to the laboratory in order to obtain samples with higher number of viable MSCs without microbiological contamination. Different biomaterials like chitosan-silicate hybrid, collagen, PLGA90:10, poly(DL-lactide-ɛ-caprolactone), and poly(vinyl alcohol) loaded with electrical conductive materials, associated to MSCs have also been tested in the rat sciatic nerve in axonotmesis and neurotmesis lesions. The in vitro studies of the scaffolds included citocompatibility evaluation of the biomaterials used and cell characterization by imunocytochemistry, karyotype analysis, differentiation capacity into neuroglial-like cells, and flow cytometry. The regeneration process follow-up has been performed by functional analysis and the repaired nerves processed for stereological studies permitted the morphologic regeneration evaluation. The MSCs from Wharton's jelly delivered through tested biomaterials should be regarded a potentially valuable tool to improve clinical outcome especially after trauma to sensory nerves. In addition, these cells represent a noncontroversial source of primitive mesenchymal progenitor cells, which can be harvested after birth, cryogenically stored, thawed, and expanded for therapeutic uses. The importance of a longitudinal study concerning tissue engineering of the peripheral nerve, which includes a multidisciplinary team able to develop biomaterials associated to cell therapies, to perform preclinical trials concerning animal welfare and the appropriate animal model is here enhanced.
Collapse
Affiliation(s)
- Jorge Ribeiro
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências e Tecnologias Agrárias e Agro-Alimentares (ICETA), Universidade do Porto (UP), Porto, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Grochmal J, Dhaliwal S, Stys PK, van Minnen J, Midha R. Skin-derived precursor Schwann cell myelination capacity in focal tibial demyelination. Muscle Nerve 2014; 50:262-72. [PMID: 24282080 DOI: 10.1002/mus.24136] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 10/29/2013] [Accepted: 11/22/2013] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Skin-derived precursor cells (SKPs) are neural crest progenitor cells that can attain a Schwann cell-like phenotype through in vitro techniques (SKP-SCs). We hypothesized that SKP-SCs could produce mature myelin and, in doing so, facilitate the recovery of a focal demyelination injury. METHODS We unilaterally injected DiI-labeled, green fluorescent protein (GFP)-producing SKP-SCs into the tibial nerves of 10 adult Lewis rats (with contralateral media control), 9 days after bilateral doxorubicin injury (0.38 μg). Tibial compound motor action potentials (CMAPs) were followed for 57 days. A separate morphometric cohort also included a Schwann cell injection group. RESULTS SKP-injected nerves recovered fastest in terms of electrophysiology and morphometry. SKP-SCs formed morphologically mature myelin, accounting for 15.3 ± 5.3% of the total myelin in SKP-SC-injected nerves. CONCLUSIONS SKP-SCs are robustly capable of myelination. They improve the recovery of a focal tibial nerve demyelination model by myelinating a measured percentage of axons.
Collapse
Affiliation(s)
- Joey Grochmal
- Department of Clinical Neuroscience, Faculty of Medicine, University of Calgary, HMRB 110-330 Hospital Drive NW, Calgary, Alberta, T2N4N1, Canada
| | | | | | | | | |
Collapse
|
33
|
Hundepool CA, Nijhuis THJ, Mohseny B, Selles RW, Hovius SER. The effect of stem cells in bridging peripheral nerve defects: a meta-analysis. J Neurosurg 2014; 121:195-209. [PMID: 24816327 DOI: 10.3171/2014.4.jns131260] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED OBJECT.: For decades the gold standard for reconstructing a large peripheral nerve defect has been, and remains, the nerve autograft. Alternatives to the nerve autograft include biological conduits and vessels. Adding stem cells in the lumen of a nerve conduit has been the subject of multiple studies. The purpose of the present meta-analysis was to summarize animal experimental studies on the effect of stem cells as a luminal additive when reconstructing a peripheral nerve defect with a nerve graft. METHODS A literature search of the MEDLINE and Embase databases was performed from inception to April 2012, searching for animal experiments on peripheral nerve reconstruction models in which a nerve conduit was used with and without the support of 3 different types of stem cells. Stem cells were analyzed according to their origin: bone marrow, adipose tissue, and other origins. Included studies had consistent outcome measurements: walking track analysis, muscle mass ratio, and electrophysiology. RESULTS Forty-four studies were included in the final analysis. Forest plots of the 3 outcome measurements (walking track analysis, muscle mass ratio, and electrophysiology) showed positive effects of stem cells on the regeneration of peripheral nerves at different time points. Almost all comparisons showed significant differences for all 3 stem cells groups compared with a control group in which stem cells were not used. CONCLUSIONS The present report systematically analyzed the different studies that used stem cells as a luminal additive when bridging a large peripheral nerve defect. All 3 different stem cell groups showed a beneficial effect when used in the reconstruction compared with control groups in which stem cells were not used.
Collapse
|
34
|
Li Y, Guo L, Ahn HS, Kim MH, Kim SW. Amniotic mesenchymal stem cells display neurovascular tropism and aid in the recovery of injured peripheral nerves. J Cell Mol Med 2014; 18:1028-34. [PMID: 24708439 PMCID: PMC4508143 DOI: 10.1111/jcmm.12249] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 01/18/2014] [Indexed: 12/15/2022] Open
Abstract
Recently, we reported that human amniotic membrane-derived mesenchymal stem cells (AMMs) possess great angiogenic potential. In this study, we determined whether local injection of AMMs ameliorates peripheral neuropathy. AMMs were transplanted into injured sciatic nerves. AMM injection promoted significant recovery of motor nerve conduction velocity and voltage amplitude compared to human adipose-derived mesenchymal stem cells. AMM implantation also augmented blood perfusion and increased intraneural vascularity. Whole-mount fluorescent imaging analysis demonstrated that AMMs exhibited higher engraftment and endothelial incorporation abilities in the sciatic nerve. In addition, the higher expression of pro-angiogenic factors was detected in AMMs injected into the peripheral nerve. Therefore, these data provide novel therapeutic and mechanistic insights into stem cell biology, and AMM transplantation may represent an alternative therapeutic option for treating peripheral neuropathy.
Collapse
Affiliation(s)
- YongNan Li
- Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | | | | | | | | |
Collapse
|
35
|
Zhao X, Yao GS, Liu Y, Wang J, Satkunendrarajah K, Fehlings M. The role of neural precursor cells and self assembling peptides in nerve regeneration. J Otolaryngol Head Neck Surg 2013; 42:60. [PMID: 24351041 PMCID: PMC3878249 DOI: 10.1186/1916-0216-42-60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 11/23/2013] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Cranial nerve injury involves loss of central neural cells in the brain stem and surrounding support matrix, leading to severe functional impairment. Therapeutically targeting cellular replacement and enhancing structural support may promote neural regeneration. We examined the combinatorial effect of neural precursor cells (NPC) and self assembling peptide (SAP) administration on nerve regeneration. METHODS Nerve injury was induced by clip compression of the rodent spinal cord. SAPs were injected immediately into the injured cord and NPCs at 2 weeks post-injury. Behavioral analysis was done weekly and rats were sacrificed at 11 weeks post injury. LFB-H&E staining was done on cord tissue to assess cavitation volume. Motor evoked potentials (MEP) were measured at week 11 to assess nerve conduction and Kaplan Meier curves were created to compare survival estimates. RESULTS NPCs and SAPs were distributed both caudal and rostral to the injury site. Behavioral analysis showed that SAP + NPC transplantation significantly improved locomotor score p <0.03) and enhanced survival (log rank test, p = 0.008) compared to control. SAP + NPC treatment also improved nerve conduction velocity (p = 0.008) but did not affect cavitation volume (p = 0.73). CONCLUSION Combinatorial NPC and SAP injection into injured nerve tissue may enhance neural repair and regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Fehlings
- Department of Genetics and Development, Toronto Western Research Institute and Spinal Program, Krembil Neuroscience Centre, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Petrova ES, Isaeva EN, Korzhevskii DE. Differentiation of dissociated rat embryonic brain after allotransplantation into damaged nerve. Bull Exp Biol Med 2013; 156:136-8. [PMID: 24319710 DOI: 10.1007/s10517-013-2296-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fragments of the dorsolateral wall of the anterior brain vesicle from rat embryos on embryonic day 15 were dissociated and the resultant suspension containing single cells and cell aggregates was injected into the proximal segment of crushed sciatic nerve of adult animals for evaluation of their engrafting and differentiation under conditions of changed microenvironment. On days 1 and 21 postoperation, Msi-1, GFAP, NeuN, vimentin, and PCNA were detected by immunohistochemical methods. Small clusters of Msi-1-immunopositive cells were detected in the nerve trunks on the next day after transplantation. On day 21 after surgery, these precursors differentiate into nerve cells, astrocytes, and primarily ependymocytes.
Collapse
Affiliation(s)
- E S Petrova
- Institute of Experimental Medicine, North-Western Branch of the Russian Academy of Medical Sciences; State Research Institute of Highly Pure Biopreparations, Federal Medical and Biological Agency of Russia, St. Petersburg, Russia.
| | | | | |
Collapse
|
37
|
Berrocal YA, Almeida VW, Gupta R, Levi AD. Transplantation of Schwann cells in a collagen tube for the repair of large, segmental peripheral nerve defects in rats. J Neurosurg 2013; 119:720-32. [DOI: 10.3171/2013.4.jns121189] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Segmental nerve defects pose a daunting clinical challenge, as peripheral nerve injury studies have established that there is a critical nerve gap length for which the distance cannot be successfully bridged with current techniques. Construction of a neural prosthesis filled with Schwann cells (SCs) could provide an alternative treatment to successfully repair these long segmental gaps in the peripheral nervous system. The object of this study was to evaluate the ability of autologous SCs to increase the length at which segmental nerve defects can be bridged using a collagen tube.
Methods
The authors studied the use of absorbable collagen conduits in combination with autologous SCs (200,000 cells/μl) to promote axonal growth across a critical size defect (13 mm) in the sciatic nerve of male Fischer rats. Control groups were treated with serum only–filled conduits of reversed sciatic nerve autografts. Animals were assessed for survival of the transplanted SCs as well as the quantity of myelinated axons in the proximal, middle, and distal portions of the channel.
Results
Schwann cell survival was confirmed at 4 and 16 weeks postsurgery by the presence of prelabeled green fluorescent protein–positive SCs within the regenerated cable. The addition of SCs to the nerve guide significantly enhanced the regeneration of myelinated axons from the nerve stump into the proximal (p < 0.001) and middle points (p < 0.01) of the tube at 4 weeks. The regeneration of myelinated axons at 16 weeks was significantly enhanced throughout the entire length of the nerve guide (p < 0.001) as compared with their number in a serum–only filled tube and was similar in number compared with the reversed autograft. Autotomy scores were significantly lower in the animals whose sciatic nerve was repaired with a collagen conduit either without (p < 0.01) or with SCs (p < 0.001) when compared with a reversed autograft.
Conclusions
The technique of adding SCs to a guidance channel significantly enhanced the gap distance that can be repaired after peripheral nerve injury with long segmental defects and holds promise in humans. Most importantly, this study represents some of the first essential steps in bringing autologous SC-based therapies to the domain of peripheral nerve injuries with long segmental defects.
Collapse
Affiliation(s)
- Yerko A. Berrocal
- 1The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Vania W. Almeida
- 1The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Ranjan Gupta
- 2Department of Orthopedic Surgery, University of California–Irvine, California
| | - Allan D. Levi
- 1The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; and
| |
Collapse
|
38
|
Nie X, Deng M, Yang M, Liu L, Zhang Y, Wen X. Axonal Regeneration and Remyelination Evaluation of Chitosan/Gelatin-Based Nerve Guide Combined with Transforming Growth Factor-β1 and Schwann Cells. Cell Biochem Biophys 2013; 68:163-72. [DOI: 10.1007/s12013-013-9683-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
39
|
Szynkaruk M, Kemp SWP, Wood MD, Gordon T, Borschel GH. Experimental and clinical evidence for use of decellularized nerve allografts in peripheral nerve gap reconstruction. TISSUE ENGINEERING PART B-REVIEWS 2012; 19:83-96. [PMID: 22924762 DOI: 10.1089/ten.teb.2012.0275] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the inherent capability for axonal regeneration, recovery following severe peripheral nerve injury remains unpredictable and often very poor. Surgeons typically use autologous nerve grafts taken from the patient's own body to bridge long nerve gaps. However, the amount of suitable nerve available from a given patient is limited, and using autologous grafts leaves the patient with scars, numbness, and other forms of donor-site morbidity. Therefore, surgeons and engineers have sought off-the-shelf alternatives to the current practice of autologous nerve grafting. Decellularized nerve allografts have recently become available as an alternative to traditional nerve autografting. In this review, we provide a critical analysis comparing the advantages and limitations of the three major experimental models of decellularized nerve allografts: cold preserved, freeze-thawed, and chemical detergent based. Current tissue engineering-based techniques to optimize decellularized nerve allografts are discussed. We also evaluate studies that supplement decellularized nerve grafts with exogenous factors such as Schwann cells, stem cells, and growth factors to both support and enhance axonal regeneration through the decellularized allografts. In examining the advantages and disadvantages of the studies of decellularized allografts, we suggest that experimental methods, including the animal model, graft length, follow-up time, and outcome measures of regenerative progress and success be consolidated. Finally, all clinical studies in which decellularized nerve allografts have been used to bridge nerve gaps in patients are reviewed.
Collapse
Affiliation(s)
- Mark Szynkaruk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
40
|
Brain-derived neurotrophic factor from bone marrow-derived cells promotes post-injury repair of peripheral nerve. PLoS One 2012; 7:e44592. [PMID: 23028564 PMCID: PMC3446933 DOI: 10.1371/journal.pone.0044592] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 08/03/2012] [Indexed: 12/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) stimulates peripheral nerve regeneration. However, the origin of BNDF and its precise effect on nerve repair have not been clarified. In this study, we examined the role of BDNF from bone marrow-derived cells (BMDCs) in post-injury nerve repair. Control and heterozygote BDNF knockout mice (BDNF+/−) received a left sciatic nerve crush using a cerebral blood clip. Especially, for the evaluation of BDNF from BMDCs, studies with bone marrow transplantation (BMT) were performed before the injury. We evaluated nerve function using a rotarod test, sciatic function index (SFI), and motor nerve conduction velocity (MNCV) simultaneously with histological nerve analyses by immunohistochemistry before and after the nerve injury until 8 weeks. BDNF production was examined by immunohistochemistry and mRNA analyses. After the nerve crush, the controls showed severe nerve dysfunction evaluated at 1 week. However, nerve function was gradually restored and reached normal levels by 8 weeks. By immunohistochemistry, BDNF expression was very faint before injury, but was dramatically increased after injury at 1 week in the distal segment from the crush site. BDNF expression was mainly co-localized with CD45 in BMDCs, which was further confirmed by the appearance of GFP-positive cells in the BMT study. Variant analysis of BDNF mRNA also confirmed this finding. BDNF+/− mice showed a loss of function with delayed histological recovery and BDNF+/+→BDNF+/− BMT mice showed complete recovery both functionally and histologically. These results suggested that the attenuated recovery of the BDNF+/− mice was rescued by the transplantation of BMCs and that BDNF from BMDCs has an essential role in nerve repair.
Collapse
|
41
|
Lee EJ, Xu L, Kim GH, Kang SK, Lee SW, Park SH, Kim S, Choi TH, Kim HS. Regeneration of peripheral nerves by transplanted sphere of human mesenchymal stem cells derived from embryonic stem cells. Biomaterials 2012; 33:7039-46. [PMID: 22795857 DOI: 10.1016/j.biomaterials.2012.06.047] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 06/22/2012] [Indexed: 12/13/2022]
Abstract
In cell therapy, the most important factor for therapeutic efficacy is the stable supply of cells with best engraftment efficiency. To meet this requirement, we have developed a culture strategy such as three-dimensional sphere of human embryonic stem cell-derived mesenchymal stem cells (hESC-MSCs) in serum-free medium. To investigate the in vivo therapeutic efficacy of hESC-MSC spheres in nerve injury model, we transected the sciatic nerve in athymic nude mice and created a 2-mm gap. Transplantation of hESC-MSC as sphere repaired the injured nerve significantly better than transplantation of hESC-MSC as suspended single cells in regard to 1) nerve conduction (sphere; 28.81 ± 3.55 vs. single cells; 18.04 ± 2.10, p < 0.05) and 2) susceptibility of nerve stimulation at low voltage (sphere; 0.38 ± 0.08 vs. single cells; 0.66 ± 0.11, p < 0.05) at 8 weeks. Recovery after sphere transplantation was near-complete when compared with the data of normal control (sphere 28.81 ± 3.55 vs normal 32.62 ± 2.85 in nerve conduction : sphere 0.38 ± 0.08 vs normal 0.36 ± 0.67 in susceptibility of nerve stimulation, no significant difference, respectively). Recovery in function of the injured nerve was well corroborated by the histologic evidence of regenerated nerve. In the mechanistic analysis, the supernatant of sphere-forming hESC-MSC contains hepatocyte growth factor and insulin-like growth factor-binding protein-1 significantly more than the supernatant of the single cells of hESC-MSC has, which might be the key factors for the improved engraftment efficiency and greater regeneration of injured peripheral nerve.
Collapse
Affiliation(s)
- Eun Ju Lee
- National Research Laboratory for Stem Cell Niche and IRICT, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rutten MJ, Janes MA, Chang IR, Gregory CR, Gregory KW. Development of a functional schwann cell phenotype from autologous porcine bone marrow mononuclear cells for nerve repair. Stem Cells Int 2012; 2012:738484. [PMID: 22792117 PMCID: PMC3388598 DOI: 10.1155/2012/738484] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 03/29/2012] [Indexed: 01/10/2023] Open
Abstract
Adult bone marrow mononuclear cells (BM-MNCs) are a potential resource for making Schwann cells to repair damaged peripheral nerves. However, many methods of producing Schwann-like cells can be laborious with the cells lacking a functional phenotype. The objective of this study was to develop a simple and rapid method using autologous BM-MNCs to produce a phenotypic and functional Schwann-like cell. Adult porcine bone marrow was collected and enriched for BM-MNCs using a SEPAX device, then cells cultured in Neurobasal media, 4 mM L-glutamine and 20% serum. After 6-8 days, the cultures expressed Schwann cell markers, S-100, O4, GFAP, were FluoroMyelin positive, but had low p75(NGF) expression. Addition of neuregulin (1-25 nM) increased p75(NGF) levels at 24-48 hrs. We found ATP dose-dependently increased intracellular calcium [Ca(2+)](i), with nucleotide potency being UTP = ATP > ADP > AMP > adenosine. Suramin blocked the ATP-induced [Ca(2+)](i) but α, β,-methylene-ATP had little effect suggesting an ATP purinergic P2Y2 G-protein-coupled receptor is present. Both the Schwann cell markers and ATP-induced [Ca(2+)](i) sensitivity decreased in cells passaged >20 times. Our studies indicate that autologous BM-MNCs can be induced to form a phenotypic and functional Schwann-like cell which could be used for peripheral nerve repair.
Collapse
Affiliation(s)
- Michael J. Rutten
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael Ann Janes
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
| | - Ivy R. Chang
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
| | - Cynthia R. Gregory
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
- Oregon Biomedical Engineering Institute, 25999 SW Canyon Creek Rd., Wilsonville, OR 97070, USA
- Portland VA Medical Center, 3710 SW U.S. Veterans Hospital Rd., Portland, OR 97239, USA
| | - Kenton W. Gregory
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
- Oregon Biomedical Engineering Institute, 25999 SW Canyon Creek Rd., Wilsonville, OR 97070, USA
| |
Collapse
|
43
|
Fibrin conduit supplemented with human mesenchymal stem cells and immunosuppressive treatment enhances regeneration after peripheral nerve injury. Neurosci Lett 2012; 516:171-6. [PMID: 22465323 DOI: 10.1016/j.neulet.2012.03.041] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 02/28/2012] [Accepted: 03/14/2012] [Indexed: 12/21/2022]
Abstract
To address the need for the development of bioengineered replacement of a nerve graft, a novel two component fibrin glue conduit was combined with human mesenchymal stem cells (MSC) and immunosupressive treatment with cyclosporine A. The effects of MSC on axonal regeneration in the conduit and reaction of activated macrophages were investigated using sciatic nerve injury model. A 10mm gap in the sciatic nerve of a rat was created and repaired either with fibrin glue conduit containing diluted fibrin matrix or fibrin glue conduit containing fibrin matrix with MSC at concentration of 80×10(6) cells/ml. Cells were labeled with PKH26 prior to transplantation. The animals received daily injections of cyclosporine A. After 3 weeks the distance of regeneration and area occupied by regenerating axons and ED1 positives macrophages was measured. MSC survived in the conduit and enhanced axonal regeneration only when transplantation was combined with cyclosporine A treatment. Moreover, addition of cyclosporine A to the conduits with transplanted MSC significantly reduced the ED1 macrophage reaction.
Collapse
|
44
|
Hsueh YY, Chiang YL, Wu CC, Lin SC. Spheroid formation and neural induction in human adipose-derived stem cells on a chitosan-coated surface. Cells Tissues Organs 2012; 196:117-28. [PMID: 22327282 DOI: 10.1159/000332045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2011] [Indexed: 12/17/2022] Open
Abstract
The application of stem cells appears to have great therapeutic potential to facilitate nerve regeneration in patients with neurodegenerative disease or spinal cord injury. Human adipose-derived stem cells (hADSCs), a subset of multipotent mesenchymal stem cells, possess the great advantages of an abundant amount of cells, less ethical conflict and minimal invasive surgical procedures to obtain the cells. Chitosan, a naturally derived polysaccharide from chitin, has been widely studied to facilitate and guide the direction of nerve regeneration as a biomaterial for the neural tube. Chitosan also serves as a three-dimensional culture substrate to facilitate cellular sphere formation among various cells but is as yet unexplored in hADSCs. In this study, the ability of hADSCs to transdifferentiate from the mesenchymal into the neural lineage by seeding hADSCs on a chitosan-coated surface to form therapeutic cell spheres was investigated. The optimal seeding density (2 × 10(4) cells/cm(2)) and harvesting time (72 h) to obtain sphere formation were determined by cell viability on a chitosan-coated surface. Expression of neural lineage markers was observed by immunofluorescent staining of nestin, neurofilament heavy chain and glial fibrillary acidic protein. The neural induction potentials were also provoked by replating spheres from primary to tertiary passages. The effect of neural induction in hADSCs on a chitosan-coated surface may help to provide cell sources for facilitating nerve regeneration in future clinical applications.
Collapse
Affiliation(s)
- Yuan-Yu Hsueh
- Division of Plastic Surgery, National Cheng Kung University, Tainan, Taiwan, ROC
| | | | | | | |
Collapse
|
45
|
Adult-brain-derived neural stem cells grafting into a vein bridge increases postlesional recovery and regeneration in a peripheral nerve of adult pig. Stem Cells Int 2012; 2012:128732. [PMID: 22448170 PMCID: PMC3289924 DOI: 10.1155/2012/128732] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/06/2011] [Accepted: 10/07/2011] [Indexed: 11/18/2022] Open
Abstract
We attempted transplantation of adult neural stem cells (ANSCs) inside an autologous venous graft following surgical transsection of nervis cruralis with 30 mm long gap in adult pig. The transplanted cell suspension was a primary culture of neurospheres from adult pig subventricular zone (SVZ) which had been labeled in vitro with BrdU or lentivirally transferred fluorescent protein. Lesion-induced loss of leg extension on the thigh became definitive in controls but was reversed by 45–90 days after neurosphere-filled vein grafting. Electromyography showed stimulodetection recovery in neurosphere-transplanted pigs but not in controls. Postmortem immunohistochemistry revealed neurosphere-derived cells that survived inside the venous graft from 10 to 240 post-lesion days and all displayed a neuronal phenotype. Newly formed neurons were distributed inside the venous graft along the severed nerve longitudinal axis. Moreover, ANSC transplantation increased CNPase expression, indicating activation of intrinsic Schwann cells. Thus ANSC transplantation inside an autologous venous graft provides an efficient repair strategy.
Collapse
|
46
|
Abstract
There are many similarities between health issues affecting military and civilian patient populations, with the exception of the relatively small but vital segment of active soldiers who experience high-energy blast injuries during combat. A rising incidence of major injuries from explosive devices in recent campaigns has further complicated treatment and recovery, highlighting the need for tissue regenerative options and intensifying interest in the possible role of stem cells for military medicine. In this review we outline the array of tissue-specific injuries typically seen in modern combat - as well as address a few complications unique to soldiers - and discuss the state of current stem cell research in addressing each area. Embryonic, induced-pluripotent and adult stem cell sources are defined, along with advantages and disadvantages unique to each cell type. More detailed stem cell sources are described in the context of each tissue of interest, including neural, cardiopulmonary, musculoskeletal and sensory tissues, with brief discussion of their potential role in regenerative medicine moving forward. Additional commentary is given to military stem cell applications aside from regenerative medicine, such as blood pharming, immunomodulation and drug screening, with an overview of stem cell banking and the unique opportunity provided by the military and civilian overlap of stem cell research.
Collapse
Affiliation(s)
- Gregory T Christopherson
- The National Institutes of Health, The National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20892, USA
| | | |
Collapse
|
47
|
Lopatina T, Kalinina N, Karagyaur M, Stambolsky D, Rubina K, Revischin A, Pavlova G, Parfyonova Y, Tkachuk V. Adipose-derived stem cells stimulate regeneration of peripheral nerves: BDNF secreted by these cells promotes nerve healing and axon growth de novo. PLoS One 2011; 6:e17899. [PMID: 21423756 PMCID: PMC3056777 DOI: 10.1371/journal.pone.0017899] [Citation(s) in RCA: 204] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 02/14/2011] [Indexed: 01/05/2023] Open
Abstract
Transplantation of adipose-derived mesenchymal stem cells (ASCs) induces tissue regeneration by accelerating the growth of blood vessels and nerve. However, mechanisms by which they accelerate the growth of nerve fibers are only partially understood. We used transplantation of ASCs with subcutaneous matrigel implants (well-known in vivo model of angiogenesis) and model of mice limb reinnervation to check the influence of ASC on nerve growth. Here we show that ASCs stimulate the regeneration of nerves in innervated mice's limbs and induce axon growth in subcutaneous matrigel implants. To investigate the mechanism of this action we analyzed different properties of these cells and showed that they express numerous genes of neurotrophins and extracellular matrix proteins required for the nerve growth and myelination. Induction of neural differentiation of ASCs enhances production of brain-derived neurotrophic factor (BDNF) as well as ability of these cells to induce nerve fiber growth. BDNF neutralizing antibodies abrogated the stimulatory effects of ASCs on the growth of nerve sprouts. These data suggest that ASCs induce nerve repair and growth via BDNF production. This stimulatory effect can be further enhanced by culturing the cells in neural differentiation medium prior to transplantation.
Collapse
Affiliation(s)
- Tatiana Lopatina
- Department of Biochemistry and Molecular Medicine, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tsai FC, Hsieh MS, Chou CM. Comparison between neurectomy and botulinum toxin A injection for denervated skeletal muscle. J Neurotrauma 2010; 27:1509-16. [PMID: 20528164 DOI: 10.1089/neu.2010.1320] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Neurectomy and botulinum toxin A (BoNT-A) injection cause denervated muscle atrophy, but questions remain about their clinical utility. We investigated time-series alterations of rat muscle weight, functional deficits, signaling pathways, and microscopic structures, to gain an understanding of the clinical implications. Between 2008 and 2009, the maximal calf circumference of patients for calf reduction either by neurectomy or BoNT-A injections were recorded for study. A rat skeletal muscle model was established through repeated or dose-adjusted BoNT-A injections and neurectomy. The survival, apoptosis pathways, functional deficits, and microscopic structures were investigated using Western blot, sciatic functional index (SFI), and transmission electron microscopy (TEM), respectively. The rat muscle weight ratio of the BoNT-A group had recovered to 89.3 +/- 3.8% by week 58, but it never recovered in the neurectomy group. Muscle weight reduction by BoNT-A not only depended on the dose, but additive effects were also obtained through repeated injections. Rat SFI demonstrated rapid recovery in both groups. Molecular expressions showed a coherent and biphasic pattern. p-Akt and apoptosis-inducing factor (AIF) were upregulated significantly, with a peak at 8 weeks in the neurectomy group (p < 0.01), but cleaved caspase-9 and caspase-3 showed no significant changes in either group. TEM findings showed irreversible and reversible inner-structure disruption and sarcomere discontinuity in the neurectomy and BoNT-A groups, respectively. We demonstrated that denervation induced lasting muscle weight and structural changes of different degrees. Muscle weight reduction by BoNT-A was related to frequency and dose. AIF-mediated caspase-independent apoptosis was significantly different for neurectomy and BoNT-A injection.
Collapse
Affiliation(s)
- Feng-Chou Tsai
- Division of Plastic Surgery, Department of Surgery, Taipei Medical University Hospital, Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | |
Collapse
|
49
|
Controlled delivery of glial cell line-derived neurotrophic factor enhances motor nerve regeneration. J Hand Surg Am 2010; 35:2008-17. [PMID: 21035963 DOI: 10.1016/j.jhsa.2010.08.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 08/13/2010] [Accepted: 08/18/2010] [Indexed: 02/02/2023]
Abstract
PURPOSE To determine the effect of a motor-specific neurotrophic factor, glial-derived neurotrophic factor (GDNF) on motor nerve regeneration. METHODS We used a nerve conduit filled with a fibrin-based delivery system that provided controlled release of GDNF during nerve regeneration. The motor branch of the rat femoral nerve was used to assess motor nerve regeneration across a 5-mm gap. Four experimental groups (n = 4 to n = 8) were evaluated. These included GDNF with the fibrin-based delivery system (GDNF-DS), fibrin alone, empty conduit (negative control), and nerve isograft (positive control). Nerves were harvested at 5 weeks for analysis by histomorphometry and electron microscopy. RESULTS At 5 mm distal to the conduit or isografts, the GDNF-DS group was not significantly different from the nerve isograft group in the following histomorphometric measures: total nerve fibers, percentage of neural tissue, and nerve density. Both the GDNF-DS and isograft groups had significantly more fibers and a higher percentage of neural tissue than fibrin alone and empty conduit groups. There were no differences in fiber width among all groups. By electron microscopy, the GDNF-DS and isograft groups also demonstrated more organized nerve architecture than the fibrin alone and empty conduit groups. CONCLUSIONS The delivery of GDNF from the fibrin-based delivery system promotes motor nerve regeneration at a level similar to an isograft in the femoral motor nerve model. This study gives insight into the potential beneficial role of GDNF in the treatment of motor nerve injuries.
Collapse
|
50
|
|