51
|
Shen YY, Gu XK, Zhang RR, Qian TM, Li SY, Yi S. Biological characteristics of dynamic expression of nerve regeneration related growth factors in dorsal root ganglia after peripheral nerve injury. Neural Regen Res 2020; 15:1502-1509. [PMID: 31997815 PMCID: PMC7059586 DOI: 10.4103/1673-5374.274343] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 06/21/2019] [Accepted: 09/20/2019] [Indexed: 01/23/2023] Open
Abstract
The regenerative capacity of peripheral nerves is limited after nerve injury. A number of growth factors modulate many cellular behaviors, such as proliferation and migration, and may contribute to nerve repair and regeneration. Our previous study observed the dynamic changes of genes in L4-6 dorsal root ganglion after rat sciatic nerve crush using transcriptome sequencing. Our current study focused on upstream growth factors and found that a total of 19 upstream growth factors were dysregulated in dorsal root ganglions at 3, 9 hours, 1, 4, or 7 days after nerve crush, compared with the 0 hour control. Thirty-six rat models of sciatic nerve crush injury were prepared as described previously. Then, they were divided into six groups to measure the expression changes of representative genes at 0, 3, 9 hours, 1, 4 or 7 days post crush. Our current study measured the expression levels of representative upstream growth factors, including nerve growth factor, brain-derived neurotrophic factor, fibroblast growth factor 2 and amphiregulin genes, and explored critical signaling pathways and biological process through bioinformatic analysis. Our data revealed that many of these dysregulated upstream growth factors, including nerve growth factor, brain-derived neurotrophic factor, fibroblast growth factor 2 and amphiregulin, participated in tissue remodeling and axon growth-related biological processes Therefore, the experiment described the expression pattern of upstream growth factors in the dorsal root ganglia after peripheral nerve injury. Bioinformatic analysis revealed growth factors that may promote repair and regeneration of damaged peripheral nerves. All animal surgery procedures were performed in accordance with Institutional Animal Care Guidelines of Nantong University and ethically approved by the Administration Committee of Experimental Animals, China (approval No. 20170302-017) on March 2, 2017.
Collapse
Affiliation(s)
- Yin-Ying Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiao-Kun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Rui-Rui Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Tian-Mei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Shi-Ying Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
52
|
Xi C, Zhang Y, Yan M, Lv Q, Lu H, Zhou J, Wang Y, Li J. Exogenous neuritin treatment improves survivability and functions of Schwann cells with improved outgrowth of neurons in rat diabetic neuropathy. J Cell Mol Med 2020; 24:10166-10176. [PMID: 32667138 PMCID: PMC7520300 DOI: 10.1111/jcmm.15627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 11/30/2022] Open
Abstract
Pathogenesis and treatment for diabetic neuropathy are still complex. A deficit of neurotrophic factors affecting Schwann cells is a very important cause of diabetic neuropathy. Neuritin is a newly discovered potential neurotrophic factor. In this study, we explored the effect of exogenous neuritin on survivability and functions of diabetic Schwann cells of rats with experimental diabetic neuropathy. Diabetic neuropathy was induced in rats. 12‐week diabetic rats contrasted with non‐diabetic normal rats had decreased levels of serum neuritin and slowed nerve conduction velocities (NCVs). Schwann cells isolated from these diabetic rats and cultured in high glucose showed reduced cell neuritin mRNA and protein and supernatant neuritin protein, increased apoptosis rates, increased caspase‐3 activities and progressively reduced viability. In contrast, exogenous neuritin treatment reduced apoptosis and improved viability, with elevated Bcl‐2 levels (not Bax) and decreased caspase‐3 activities. Co‐cultured with diabetic Schwann cells pre‐treated with exogenous neuritin in high glucose media, and diabetic DRG neurons showed lessened decreased neurite outgrowth and supernatant NGF concentration occurring in co‐culture of diabetic cells. Exogenous neuritin treatment ameliorated survivability and functions of diabetic Schwann cells of rats with diabetic neuropathy. Our study may provide a new mechanism and potential treatment for diabetic neuropathy.
Collapse
Affiliation(s)
- Chunhong Xi
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yingduan Zhang
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mei Yan
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qing Lv
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huan Lu
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Zhou
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yucheng Wang
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianbo Li
- Endocrinology and Metabolism Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Diabetic Neuropathy Study Group of Chinese Diabetes Society, Beijing, China
| |
Collapse
|
53
|
Yang S, Wang C, Zhu J, Lu C, Li H, Chen F, Lu J, Zhang Z, Yan X, Zhao H, Sun X, Zhao L, Liang J, Wang Y, Peng J, Wang X. Self-assembling peptide hydrogels functionalized with LN- and BDNF- mimicking epitopes synergistically enhance peripheral nerve regeneration. Theranostics 2020; 10:8227-8249. [PMID: 32724468 PMCID: PMC7381722 DOI: 10.7150/thno.44276] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
The regenerative capacity of the peripheral nervous system is closely related to the role that Schwann cells (SCs) play in construction of the basement membrane containing multiple extracellular matrix proteins and secretion of neurotrophic factors, including laminin (LN) and brain-derived neurotrophic factor (BDNF). Here, we developed a self-assembling peptide (SAP) nanofiber hydrogel based on self-assembling backbone Ac-(RADA)4-NH2 (RAD) dual-functionalized with laminin-derived motif IKVAV (IKV) and a BDNF-mimetic peptide epitope RGIDKRHWNSQ (RGI) for peripheral nerve regeneration, with the hydrogel providing a three-dimensional (3D) microenvironment for SCs and neurites. Methods: Circular dichroism (CD), atomic force microscopy (AFM), and scanning electron microscopy (SEM) were used to characterize the secondary structures, microscopic structures, and morphologies of self-assembling nanofiber hydrogels. Then the SC adhesion, myelination and neurotrophin secretion were evaluated on the hydrogels. Finally, the SAP hydrogels were injected into hollow chitosan tubes to bridge a 10-mm-long sciatic nerve defect in rats, and in vivo gene expression at 1 week, axonal regeneration, target muscular re-innervation, and functional recovery at 12 weeks were assessed. Results: The bioactive peptide motifs were covalently linked to the C-terminal of the self-assembling peptide and the functionalized peptides could form well-defined nanofibrous hydrogels capable of providing a 3D microenvironment similar to native extracellular matrix. SCs displayed improved cell adhesion on hydrogels with both IKV and RGI, accompanied by increased cell spreading and elongation relative to other groups. RSCs cultured on hydrogels with IKV and RGI showed enhanced gene expression of NGF, BDNF, CNTF, PMP22 and NRP2, and decreased gene expression of NCAM compared with those cultured on other three groups after a 7-day incubation. Additionally, the secretion of NGF, BDNF, and CNTF of RSCs was significantly improved on dual-functionalized peptide hydrogels after 3 days. At 1 week after implantation, the expressions of neurotrophin and myelin-related genes in the nerve grafts in SAP and Autograft groups were higher than that in Hollow group, and the expression of S100 in groups containing both IKV and RGI was significantly higher than that in groups containing either IKV or RGI hydrogels, suggesting enhanced SC proliferation. The morphometric parameters of the regenerated nerves, their electrophysiological performance, the innervated muscle weight and remodeling of muscle fibers, and motor function showed that RAD/IKV/RGI and RAD/IKV-GG-RGI hydrogels could markedly improve axonal regeneration with enhanced re-myelination and motor functional recovery through the synergetic effect of IKV and RGI functional motifs. Conclusions: We found that the dual-functionalized SAP hydrogels promoted RSC adhesion, myelination, and neurotrophin secretion in vitro and successfully bridged a 10-mm gap representing a sciatic nerve defect in rats in vivo. The results demonstrated the synergistic effect of IKVAV and RGI on axonal regrowth and function recovery after peripheral nerve injury.
Collapse
Affiliation(s)
- Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Chong Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jinjin Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang, Hangzhou 310016, China
| | - Changfeng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing 100191, China
| | - Haitao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Fuyu Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jiaju Lu
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaoqing Yan
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- School of Clinical Medicine, Tsinghua University, Beijing 100084, China
| | - He Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jing Liang
- Department of Pediatrics, Tianjin Hospital, Tianjin University, No. 406 Jiefang Nan Road, Tianjin 300211, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226007, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
54
|
Ju C, Park E, Kim T, Kim T, Kang M, Lee KS, Park SM. Effectiveness of electrical stimulation on nerve regeneration after crush injury: Comparison between invasive and non-invasive stimulation. PLoS One 2020; 15:e0233531. [PMID: 32453807 PMCID: PMC7250463 DOI: 10.1371/journal.pone.0233531] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/06/2020] [Indexed: 01/09/2023] Open
Abstract
Several studies have investigated the use of invasive and non-invasive stimulation methods to enhance nerve regeneration, and varying degrees of effectiveness have been reported. However, due to the use of different parameters in these studies, a fair comparison between the effectiveness of invasive and non-invasive stimulation methods is not possible. The present study compared the effectiveness of invasive and non-invasive stimulation using similar parameters. Eighteen Sprague Dawley rats were classified into three groups: the iES group stimulated with fully implantable device, the tES group stimulated with transcutaneous electrical nerve stimulation (TENS), and the injury group (no stimulation). The iES and tES groups received stimulation for 6 weeks starting immediately after the injury. Motor function was evaluated using the sciatic functional index (SFI) every week. The SFI values increased over time in all groups; faster and superior functional recovery was observed in the iES group than in the tES group. Histological evaluation of the nerve sections and gastrocnemius muscle sections were performed every other week. The axon diameter and muscle fiber area in the iES group were larger, and the g-ratio in the iES group was closer to 0.6 than those in the tES group. To assess the cause of the difference in efficiency, a 3D rat anatomical model was used to simulate the induced electric fields in each group. A significantly higher concentration and intensity around the sciatic nerve was observed in the iES group than in the tES group. Vector field distribution showed that the field was orthogonal to the sciatic nerve spread in the tES group, whereas it was parallel in the iES group; this suggested that the tES group was less effective in nerve stimulation. The results indicated that even though rats in the TENS group showed better recovery than those in the injury group, it cannot replace direct stimulation yet because rats stimulated with the invasive method showed faster recovery and superior outcomes. This was likely attributable to the greater concentration and parallel distribution of electric field with respect to target nerve.
Collapse
Affiliation(s)
- Chanyang Ju
- Department of Creative IT Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Eunkyoung Park
- Biomedical Engineering Research Center, Smart Healthcare Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Taewoo Kim
- Biomedical Engineering Research Center, Smart Healthcare Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Taekyung Kim
- Biomedical Engineering Research Center, Smart Healthcare Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Minhee Kang
- Biomedical Engineering Research Center, Smart Healthcare Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyu-Sung Lee
- Biomedical Engineering Research Center, Smart Healthcare Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- * E-mail: (KSL); (SMP)
| | - Sung-Min Park
- Department of Creative IT Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- * E-mail: (KSL); (SMP)
| |
Collapse
|
55
|
Labroo P, Ho S, Sant H, Shea JE, Agarwal J, Gale B. Modeling diffusion-based drug release inside a nerve conduit in vitro and in vivo validation study. Drug Deliv Transl Res 2020; 11:154-168. [PMID: 32367424 DOI: 10.1007/s13346-020-00755-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The objective of this work was to develop a model and understand the diffusion of a drug into and throughout a drug delivering nerve conduit from a surrounding reservoir through a hole in the wall separating the lumen of the conduit and the reservoir. A mathematical model based on Fick's law of diffusion was developed using the finite difference method to understand the drug diffusion and the effect of varying device parameters on the concentration of drug delivered from a hole-based drug delivery device. The mathematical model was verified using a physical microfluidic (μFD) model and an in vitro/in vivo release test using prototype devices. The results of the mathematical model evaluation and microfluidic device testing offered positive insight into the reliability and function of the reservoir and hole-based drug delivering nerve conduit. The mathematical model demonstrated how changing device parameters would change the drug concentration inside the device. It was observed that the drug release in the conduit could be tuned by both concentration scaling and changing the hole size or number of holes. Based on the results obtained from the microfluidic device, the error in the mathematical drug release model was shown to be less than 10% when comparing the data obtained from mathematical model and μFD model. The data highlights the flexibility of having a hole-based drug delivery system, since the drug release can be scaled predictably by changing the device parameters or the concentration of the drug in the reservoir. Graphical abstract .
Collapse
Affiliation(s)
- Pratima Labroo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Scott Ho
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Himanshu Sant
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jill E Shea
- Department of Surgery, University of Utah, 30 N 1900 E, 3b400, Salt Lake City, UT, 84112-9057, USA
| | - Jayant Agarwal
- Department of Surgery, University of Utah, 30 N 1900 E, 3b400, Salt Lake City, UT, 84112-9057, USA.
| | - Bruce Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
56
|
Zhou G, Chang W, Zhou X, Chen Y, Dai F, Anwar A, Yu X. Nanofibrous Nerve Conduits with Nerve Growth Factors and Bone Marrow Stromal Cells Pre-Cultured in Bioreactors for Peripheral Nerve Regeneration. ACS APPLIED MATERIALS & INTERFACES 2020; 12:16168-16177. [PMID: 32182427 DOI: 10.1021/acsami.0c04191] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Peripheral nerve injury (PNI) was the leading cause of permanent dysfunction in movement and sensation. Synthesized nerve guide conduits (NGCs) with Schwann Cells (SCs) can help peripheral nerve regeneration. However, poor accessibility of SCs and lack of full coverage of seeded cells on NGCs can lead to failure of nerve regeneration across long gaps and full functional recovery. To overcome these limitations, bone marrow stromal cells (BMSCs) and a novel culture method were proposed in the current study. BMSCs were harvested and seeded on a never growth factor (NGF)-loaded PCL nanofibrous NGCs and cultured with a rotary cell culture system (RCCS) before implantation. The NGCs were tested in vitro with PC-12 cells to validate the bioactivity of released NGF and to access its ability to promote neurite extension. Also, the NGCs were tested in vivo with rat sciatic nerve model to exam its potential in bridging the long gap (15 mm segmental defect). The efficacy of the NGCs was investigated based on the results of the functional test, electrophysiology test, muscle atrophy, and histological analysis. The results of in vitro PC-12 cell study confirmed the bioactivity of released NGF and showed a significant increase in the neurite extension with the help of PEG-diamine and BSA. These results showed that the novel loading method could preserve the bioactivity of growth factors and achieve a sustained release in vitro. Besides, the results of the in vivo study exhibited a significant increase with the combination of all additives. These results showed that with the help of NGF and RCCS, the NGCs with the seeded BMSCs could enhance peripheral nerve regeneration across long nerve injury gaps.
Collapse
Affiliation(s)
- Gan Zhou
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Wei Chang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Xiaqing Zhou
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Yifan Chen
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Futao Dai
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Aneela Anwar
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Xiaojun Yu
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| |
Collapse
|
57
|
Invited review: Utilizing peripheral nerve regenerative elements to repair damage in the CNS. J Neurosci Methods 2020; 335:108623. [DOI: 10.1016/j.jneumeth.2020.108623] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/20/2022]
|
58
|
Three-Dimensional Model of Dorsal Root Ganglion Explant as a Method of Studying Neurotrophic Factors in Regenerative Medicine. Biomedicines 2020; 8:biomedicines8030049. [PMID: 32138155 PMCID: PMC7175199 DOI: 10.3390/biomedicines8030049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/21/2020] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
Neurotrophic factors play a key role in the development, differentiation, and survival of neurons and nerve regeneration. In the present study, we evaluated the effect of certain neurotrophic factors (NGF, BDNF, and GDNF) on axon growth and migration of Nestin-green fluorescent protein (GFP)-positive cells using a 3D model of dorsal root ganglion (DRG) explant culture in Matrigel. Our method generally represents a convenient model for assessing the effects of soluble factors and therapeutic agents on axon growth and nerve regeneration in R&D studies. By analyzing the DRG explants in ex vivo culture for 21 days, one can evaluate the parameters of neurite outgrowth and the rate of cell migration from the DRG explants into the Matrigel. For the current study, we used Nestin-GFP-expressing mice in which neural precursors express Nestin and the green fluorescent protein (GFP) under the same promoter. We revealed that GDNF significantly (two fold) stimulated axon outgrowth (p < 0.05), but not BDNF or NGF. It is well-known that axon growth can be stimulated by activated glial cells that fulfill a trophic function for regenerating nerves. For this reason, we evaluated the number of Nestin-GFP-positive cells that migrated from the DRG into the Matrigel in our 3D ex vivo explant model. We found that NGF and GDNF, but not BDNF, stimulated the migration of Nestin-GFP cells compared to the control (p < 0.05). On the basis of the aforementioned finding, we concluded that GDNF had the greatest stimulating potential for axon regeneration, as it stimulated not only the axon outgrowth, but also glial cell migration. Although NGF significantly stimulated glial cell migration, its effect on axon growth was insufficient for axon regeneration.
Collapse
|
59
|
Yang M, Sun JY, Ying CC, Wang Y, Guo YL. Adipose-derived stem cells modified by BDNF gene rescue erectile dysfunction after cavernous nerve injury. Neural Regen Res 2020; 15:120-127. [PMID: 31535660 PMCID: PMC6862402 DOI: 10.4103/1673-5374.264464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cavernous nerve injury is the main cause of erectile dysfunction following radical prostatectomy. The recovery of erectile function following radical prostatectomy remains challenging. Our previous studies found that injecting adipose-derived stem cells (ADSCs) into the cavernosa could repair the damaged cavernous nerves, but the erectile function of the treated rats could not be restored to a normal level. We evaluated the efficacy of ADSCs infected with a lentiviral vector encoding rat brain-derived neurotrophic factor (lenti-rBDNF) in a rat model of cavernous nerve injury. The rats were equally and randomly divided into four groups. In the control group, bilateral cavernous nerves were isolated but not injured. In the bilateral cavernous nerve injury group, bilateral cavernous nerves were isolated and injured with a hemostat clamp for 2 minutes. In the ADSCGFP and ADSCrBDNF groups, after injury with a hemostat clamp for 2 minutes, rats were injected with ADSCs infected with lenti-GFP (1 × 106 in 20 μL) and lenti-rBDNF (1 × 106 in 20 μL), respectively. Erectile function was assessed 4 weeks after injury by measuring intracavernosal pressures. Then, penile tissues were collected for histological detection and western blot assay. Results demonstrated that compared with the bilateral cavernous nerve injury group, erectile function was significantly recovered in the ADSCGFP and ADSCrBDNF groups, and to a greater degree in the ADSCrBDNF group. Neuronal nitric oxide synthase content in the dorsal nerves and the ratio of smooth muscle/collagen were significantly higher in the ADSCrBDNF and ADSCGFP groups than in the bilateral cavernous nerve injury group. Neuronal nitric oxide synthase expression was obviously higher in the ADSCrBDNF group than in the ADSCGFP group. These findings confirm that intracavernous injection with ADSCs infected with lenti-rBDNF can effectively improve erectile dysfunction caused by cavernous nerve injury. This study was approved by the Medical Animal Care and Welfare Committee of Wuhan University, China (approval No. 2017-1638) on June 20, 2017.
Collapse
Affiliation(s)
- Mei Yang
- Department of Endocrinology, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei Province, China
| | - Jiang-Yang Sun
- Department of Hepatobiliary Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Cheng-Cheng Ying
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yong Wang
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yong-Lian Guo
- Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
60
|
Cheng XQ, Liang XZ, Wei S, Ding X, Han GH, Liu P, Sun X, Quan Q, Tang H, Zhao Q, Shang AJ, Peng J. Protein microarray analysis of cytokine expression changes in distal stumps after sciatic nerve transection. Neural Regen Res 2020; 15:503-511. [PMID: 31571662 PMCID: PMC6921340 DOI: 10.4103/1673-5374.266062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A large number of chemokines, cytokines, other trophic factors and the extracellular matrix molecules form a favorable microenvironment for peripheral nerve regeneration. This microenvironment is one of the major factors for regenerative success. Therefore, it is important to investigate the key molecules and regulators affecting nerve regeneration after peripheral nerve injury. However, the identities of specific cytokines at various time points after sciatic nerve injury have not been determined. The study was performed by transecting the sciatic nerve to establish a model of peripheral nerve injury and to analyze, by protein microarray, the expression of different cytokines in the distal nerve after injury. Results showed a large number of cytokines were up-regulated at different time points post injury and several cytokines, e.g., ciliary neurotrophic factor, were downregulated. The construction of a protein-protein interaction network was used to screen how the proteins interacted with differentially expressed cytokines. Kyoto Encyclopedia of Genes and Genomes pathway and Gene ontology analyses indicated that the differentially expressed cytokines were significantly associated with chemokine signaling pathways, Janus kinase/signal transducers and activators of transcription, phosphoinositide 3-kinase/protein kinase B, and notch signaling pathway. The cytokines involved in inflammation, immune response and cell chemotaxis were up-regulated initially and the cytokines involved in neuronal apoptotic processes, cell-cell adhesion, and cell proliferation were up-regulated at 28 days after injury. Western blot analysis showed that the expression and changes of hepatocyte growth factor, glial cell line-derived neurotrophic factor and ciliary neurotrophic factor were consistent with the results of protein microarray analysis. The results provide a comprehensive understanding of changes in cytokine expression and changes in these cytokines and classical signaling pathways and biological functions during Wallerian degeneration, as well as a basis for potential treatments of peripheral nerve injury. The study was approved by the Institutional Animal Care and Use Committee of the Chinese PLA General Hospital, China (approval number: 2016-x9-07) in September 2016.
Collapse
Affiliation(s)
- Xiao-Qing Cheng
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xue-Zhen Liang
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing; The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Shuai Wei
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xiao Ding
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Gong-Hai Han
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ping Liu
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xun Sun
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qi Quan
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - He Tang
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qing Zhao
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ai-Jia Shang
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
61
|
Hussain G, Wang J, Rasul A, Anwar H, Qasim M, Zafar S, Aziz N, Razzaq A, Hussain R, de Aguilar JLG, Sun T. Current Status of Therapeutic Approaches against Peripheral Nerve Injuries: A Detailed Story from Injury to Recovery. Int J Biol Sci 2020; 16:116-134. [PMID: 31892850 PMCID: PMC6930373 DOI: 10.7150/ijbs.35653] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/22/2019] [Indexed: 12/14/2022] Open
Abstract
Peripheral nerve injury is a complex condition with a variety of signs and symptoms such as numbness, tingling, jabbing, throbbing, burning or sharp pain. Peripheral nerves are fragile in nature and can easily get damaged due to acute compression or trauma which may lead to the sensory and motor functions deficits and even lifelong disability. After lesion, the neuronal cell body becomes disconnected from the axon's distal portion to the injury site leading to the axonal degeneration and dismantlement of neuromuscular junctions of targeted muscles. In spite of extensive research on this aspect, complete functional recovery still remains a challenge to be resolved. This review highlights detailed pathophysiological events after an injury to a peripheral nerve and the associated factors that can either hinder or promote the regenerative machinery. In addition, it throws light on the available therapeutic strategies including supporting therapies, surgical and non-surgical interventions to ameliorate the axonal regeneration, neuronal survival, and reinnervation of peripheral targets. Despite the availability of various treatment options, we are still lacking the optimal treatments for a perfect and complete functional regain. The need for the present age is to discover or design such potent compounds that would be able to execute the complete functional retrieval. In this regard, plant-derived compounds are getting more attention and several recent reports validate their remedial effects. A plethora of plants and plant-derived phytochemicals have been suggested with curative effects against a number of diseases in general and neuronal injury in particular. They can be a ray of hope for the suffering individuals.
Collapse
Affiliation(s)
- Ghulam Hussain
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000 Pakistan
| | - Jing Wang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian Province, 361021 China
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, 38000 Pakistan
| | - Haseeb Anwar
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000 Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, 38000 Pakistan
| | - Shamaila Zafar
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000 Pakistan
| | - Nimra Aziz
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000 Pakistan
| | - Aroona Razzaq
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, 38000 Pakistan
| | - Rashad Hussain
- Department of Neurosurgery, Center for Translational Neuromedicine (SMD), School of Medicine and Dentistry, University of Rochester Medical Center, 601 Elmwood Ave, Box 645, Rochester, NY 14642, USA
| | - Jose-Luis Gonzalez de Aguilar
- Université de Strasbourg, UMR_S 1118, Strasbourg, France
- INSERM, U1118, Mécanismes Centraux et Péripheriques de la Neurodégénérescence, Strasbourg, France
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian Province, 361021 China
| |
Collapse
|
62
|
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: 46] [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
|
63
|
Liao CF, Chen CC, Lu YW, Yao CH, Lin JH, Way TD, Yang TY, Chen YS. Effects of endogenous inflammation signals elicited by nerve growth factor, interferon-γ, and interleukin-4 on peripheral nerve regeneration. J Biol Eng 2019; 13:86. [PMID: 31754373 PMCID: PMC6854735 DOI: 10.1186/s13036-019-0216-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022] Open
Abstract
Background Large gap healing is a difficult issue in the recovery of peripheral nerve injury. The present study provides in vivo trials of silicone rubber chambers filled with collagen containing IFN-γ or IL-4 to bridge a 15 mm sciatic nerve defect in rats. Fillings of NGF and normal saline were used as the positive and negative controls. Neuronal electrophysiology, neuronal connectivity, macrophage infiltration, location and expression levels of calcitonin gene-related peptide and histology of the regenerated nerves were evaluated. Results At the end of 6 weeks, animals from the groups of NGF and IL-4 had dramatic higher rates of successful regeneration (100 and 80%) across the wide gap as compared to the groups of IFN-γ and saline controls (30 and 40%). In addition, the NGF group had significantly higher NCV and shorter latency compared to IFN-γ group (P < 0.05). The IL-4 group recruited significantly more macrophages in the nerves as compared to the saline controls and the NGF-treated animals (P < 0.05). Conclusions The current study demonstrated that NGF and IL-4 show potential growth-promoting capability for peripheral nerve regeneration. These fillings in the bridging conduits may modulate local inflammatory conditions affecting recovery of the nerves.
Collapse
Affiliation(s)
- Chien-Fu Liao
- 1Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chung-Chia Chen
- Linsen Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan
| | - Yu-Wen Lu
- 3Department of Chinese Medicine, Show Chwan Memorial Hospital, Chunaghua, Taiwan.,4Department of Chinese Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Chun-Hsu Yao
- 5Lab of Biomaterials, School of Chinese Medicine, China Medical University , Taichung, Taiwan.,6Biomaterials Translational Research Center, China Medical University Hospital, Taichung, Taiwan.,7Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jia-Horng Lin
- 8Department of Fiber and Composite Materials, Feng Chia University, Taichung, Taiwan
| | - Tzong-Der Way
- 1Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Tse-Yen Yang
- 9Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.,10Center for General Education & Master Program of Digital Health Innovation, China Medical University, Taichung, Taiwan
| | - Yueh-Sheng Chen
- 5Lab of Biomaterials, School of Chinese Medicine, China Medical University , Taichung, Taiwan.,6Biomaterials Translational Research Center, China Medical University Hospital, Taichung, Taiwan.,7Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan.,10Center for General Education & Master Program of Digital Health Innovation, China Medical University, Taichung, Taiwan.,11College of Humanities and Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
64
|
Cui W, Liu CX, Wang J, Zhang YC, Shen Q, Feng ZH, Wu J, Li JX. An oleanolic acid derivative reduces denervation-induced muscle atrophy via activation of CNTF-mediated JAK2/STAT3 signaling pathway. Eur J Pharmacol 2019; 861:172612. [PMID: 31421088 DOI: 10.1016/j.ejphar.2019.172612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/10/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Denervation caused by sciatic nerve injury has brought great harm to the patients, especially denervation-induced muscle atrophy. The body stress produces a large number of Schwann cells when the sciatic nerve is injured, and the cells secrete some cytokines including ciliary neurotrophic factor (CNTF) that not only play a role in promoting the repair of sciatic nerve, but also maintain the normal physiological function of the muscles surrounding the damaged nerves. CNTF upregulates janus kinase 2 (JAK2) and signal transducers and activators of transcription 3 (STAT3) signals in myoblasts, and consequently accelerates the proliferation and differentiation of myoblasts. This effect on myoblasts is the most effective way to relieve muscle atrophy. Therefore, increasing CNTF is a promising direction to improve muscle atrophy. In the present study, an oleanolic acid derivative, HA-19, increased the proliferation of Schwann cells, and elevated CNTF production of the cells. HA-19 up-regulated the phosphorylation of JAK2 and STAT3 not only by directly acting on myoblasts, but also by increasing the secretion of CNTF of Schwann cells; and consequently, promoted the proliferation and differentiation of myoblasts. In denervation-induced muscle atrophy mice model, treatment with HA-19 significantly increased the weights of tibialis anterior (TA), gastrocnemius (Gastroc.), extensor digitorum longus (EDL), soleus and quadriceps (Quad.) under atrophied state. And, very interestingly, these muscles under normal condition were also strengthened by HA-19. Our finding demonstrated that HA-19 has a great potential as a lead compound for the drug discovery of anti-denervation-induced muscle atrophy.
Collapse
Affiliation(s)
- Wei Cui
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Chen-Xi Liu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jie Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yu-Chao Zhang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qi Shen
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhen-Hua Feng
- The Center of Diagnosis and Treatment for Joint Disease, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, 210008, China
| | - Jing Wu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| | - Jian-Xin Li
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
65
|
Ruven C, Badea SR, Wong WM, Wu W. Combination Treatment With Exogenous GDNF and Fetal Spinal Cord Cells Results in Better Motoneuron Survival and Functional Recovery After Avulsion Injury With Delayed Root Reimplantation. J Neuropathol Exp Neurol 2019; 77:325-343. [PMID: 29420729 DOI: 10.1093/jnen/nly009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
When spinal roots are torn off from the spinal cord, both the peripheral and central nervous system get damaged. As the motoneurons lose their axons, they start to die rapidly, whereas target muscles atrophy due to the denervation. In this kind of complicated injury, different processes need to be targeted in the search for the best treatment strategy. In this study, we tested glial cell-derived neurotrophic factor (GDNF) treatment and fetal lumbar cell transplantation for their effectiveness to prevent motoneuron death and muscle atrophy after the spinal root avulsion and delayed reimplantation. Application of exogenous GDNF to injured spinal cord greatly prevented the motoneuron death and enhanced the regeneration and axonal sprouting, whereas no effect was seen on the functional recovery. In contrast, cell transplantation into the distal nerve did not affect the host motoneurons but instead mitigated the muscle atrophy. The combination of GDNF and cell graft reunited the positive effects resulting in better functional recovery and could therefore be considered as a promising strategy for nerve and spinal cord injuries that involve the avulsion of spinal roots.
Collapse
Affiliation(s)
- Carolin Ruven
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | - Wai-Man Wong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wutian Wu
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Re-Stem Biotechnology Co., Ltd, Jiangsu, China
| |
Collapse
|
66
|
Wang J, Zheng W, Chen L, Zhu T, Shen W, Fan C, Wang H, Mo X. Enhancement of Schwann Cells Function Using Graphene-Oxide-Modified Nanofiber Scaffolds for Peripheral Nerve Regeneration. ACS Biomater Sci Eng 2019; 5:2444-2456. [DOI: 10.1021/acsbiomaterials.8b01564] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Juan Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Wei Zheng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Liang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Tonghe Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Wei Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey 07030, United States
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
67
|
Günter C, Delbeke J, Ortiz-Catalan M. Safety of long-term electrical peripheral nerve stimulation: review of the state of the art. J Neuroeng Rehabil 2019; 16:13. [PMID: 30658656 PMCID: PMC6339286 DOI: 10.1186/s12984-018-0474-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/11/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Electrical stimulation of peripheral nerves is used in a variety of applications such as restoring motor function in paralyzed limbs, and more recently, as means to provide intuitive sensory feedback in limb prostheses. However, literature on the safety requirements for stimulation is scarce, particularly for chronic applications. Some aspects of nerve interfacing such as the effect of stimulation parameters on electrochemical processes and charge limitations have been reviewed, but often only for applications in the central nervous system. This review focuses on the safety of electrical stimulation of peripheral nerve in humans. METHODS We analyzed early animal studies evaluating damage thresholds, as well as more recent investigations in humans. Safety requirements were divided into two main categories: passive and active safety. We made the distinction between short-term (< 30 days) and chronic (> 30 days) applications, as well as between electrode preservation (biostability) and body tissue healthy survival (harmlessness). In addition, transferability of experimental results between different tissues and species was considered. RESULTS At present, extraneural electrodes have shown superior long-term stability in comparison to intraneural electrodes. Safety limitations on pulse amplitude (and consequently, charge injection) are dependent on geometrical factors such as electrode placement, size, and proximity to the stimulated fiber. In contrast, other parameters such as stimulation frequency and percentage of effective stimulation time are more generally applicable. Currently, chronic stimulation at frequencies below 30 Hz and percentages of effective stimulation time below 50% is considered safe, but more precise data drawn from large databases are necessary. Unfortunately, stimulation protocols are not systematically documented in the literature, which limits the feasibility of meta-analysis and impedes the generalization of conclusions. We therefore propose a standardized list of parameters necessary to define electrical stimulation and allow future studies to contribute to meta-analyses. CONCLUSION The safety of chronic continuous peripheral nerve stimulation at frequencies higher than 30 Hz has yet to be documented. Precise parameter values leading to stimulation-induced depression of neuronal excitability (SIDNE) and neuronal damage, as well as the transition between the two, are still lacking. At present, neural damage mechanisms through electrical stimulation remain obscure.
Collapse
Affiliation(s)
- Clara Günter
- Biomechatronics and Neurorehabilitation Laboratory, Department of Electrical Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden
| | - Jean Delbeke
- LCEN3, Department of Neurology, Institute of Neuroscience, Ghent University, C. Heymanslaan, 10, 9000, Ghent, Belgium
| | - Max Ortiz-Catalan
- Biomechatronics and Neurorehabilitation Laboratory, Department of Electrical Engineering, Chalmers University of Technology, 41296, Gothenburg, Sweden. .,Integrum AB, Krokslätts Fabriker 50, 43137, Mölndal, Sweden.
| |
Collapse
|
68
|
Wang J, Cheng Y, Chen L, Zhu T, Ye K, Jia C, Wang H, Zhu M, Fan C, Mo X. In vitro and in vivo studies of electroactive reduced graphene oxide-modified nanofiber scaffolds for peripheral nerve regeneration. Acta Biomater 2019; 84:98-113. [PMID: 30471474 DOI: 10.1016/j.actbio.2018.11.032] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 11/18/2018] [Accepted: 11/20/2018] [Indexed: 12/30/2022]
Abstract
Graphene, as a promising biomaterial, has received great attention in biomedical fields due to its intriguing properties, especially the conductivity and biocompatibility. Given limited studies on the effects of graphene-based scaffolds on peripheral nerve regeneration in vitro and in vivo under electrical stimulation (ES), the present study was intended to systematically investigate how conductive graphene-based nanofibrous scaffolds regulate Schwann cell (SC) behavior including migration, proliferation and myelination, and PC12 cell differentiation in vitro via ES, and whether these conductive scaffolds could guide SC migration and promote nerve regeneration in vivo. Briefly, the reduced graphene oxide (RGO) was coated onto ApF/PLCL nanofibrous scaffolds via in situ redox reaction of the graphene oxide (GO). In vitro, RGO-coated ApF/PLCL (AP/RGO) scaffolds significantly enhanced SC migration, proliferation, and myelination including myelin-specific gene expression and neurotrophic factor secretion. The conditioned media of SCs cultured on AP/RGO scaffolds under ES could induce the differentiation of PC12 cells in a separate culture. In addition, PC12 cells cultured on the conductive AP/RGO scaffolds also showed elevated differentiation upon ES. In vivo implantation of the conductive AP/RGO nerve guidance conduits into rat sciatic nerve defects exhibited a similar healing capacity to autograft, which is the current gold standard in peripheral nerve regeneration. In view of the performance of AP/RGO scaffolds in modulating cell functions in vitro and promoting nerve regeneration in vivo, it is expected that the graphene-based conductive nanofibrous scaffolds would exhibit their potential in peripheral nerve repair and regeneration. STATEMENT OF SIGNIFICANCE: Despite the demonstrated capability of bridging the distal and proximal peripheral nerves, it remains a significant challenge with current artificial nerve conduits to achieve the desired physiological functions, e.g., the transmission of electrical stimuli. Herein, we explored the possibility of combining the conductive properties of graphene with electrospun nanofiber to create the electroactive biomimetic scaffolds for nerve tissue regeneration. In vitro and in vivo studies were carried out: (1) In vitro, the conductive nanofibrous scaffolds significantly promoted SC migration, proliferation and myelination including myelin specific gene expression and neurotrophicfactor secretion, and induced PC12 cell differentiation with electrical stimulation. (2) In vivo, the conductive nerve guidance conduit exhibited similar effects with the gold standard autograft. In view of the performance of this conductive scaffold in modulating the cell functions in vitro and promoting nerve regeneration in vivo, it is expected that the graphene-modified nanofibrous scaffolds will exhibit their potential in peripheral nerve repair and regeneration.
Collapse
Affiliation(s)
- Juan Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Yuan Cheng
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liang Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Tonghe Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Kaiqiang Ye
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Chao Jia
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| |
Collapse
|
69
|
Liu QY, Miao Y, Wang XH, Wang P, Cheng ZC, Qian TM. Increased levels of miR-3099 induced by peripheral nerve injury promote Schwann cell proliferation and migration. Neural Regen Res 2019; 14:525-531. [PMID: 30539823 PMCID: PMC6334613 DOI: 10.4103/1673-5374.245478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs (miRNAs) can regulate the modulation of the phenotype of Schwann cells. Numerous novel miRNAs have been discovered and identified in rat sciatic nerve segments, including miR-3099. In the current study, miR-3099 expression levels following peripheral nerve injury were measured in the proximal stumps of rat sciatic nerves after surgical crush. Real-time reverse transcription-polymerase chain reaction was used to determine miR-3099 expression in the crushed nerve segment at 0, 1, 4, 7, and 14 days post sciatic nerve injury, which was consistent with Solexa sequencing outcomes. Expression of miR-3099 was up-regulated following peripheral nerve injury. EdU and transwell chamber assays were used to observe the effect of miR-3099 on Schwann cell proliferation and migration. The results showed that increased miR-3099 expression promoted the proliferation and migration of Schwann cells. However, reduced miR-3099 expression suppressed the proliferation and migration of Schwann cells. The potential target genes of miR-3099 were also investigated by bioinformatic tools and high-throughput outcomes. miR-3099 targets genes Aqp4, St8sia2, Tnfsf15, and Zbtb16 and affects the proliferation and migration of Schwann cells. This study examined the levels of miR-3099 at different time points following peripheral nerve injury. Our results confirmed that increased miR-3099 level induced by peripheral nerve injury can promote the proliferation and migration of Schwann cells.
Collapse
Affiliation(s)
- Qian-Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Yang Miao
- Department of Pharmacy, Yancheng City No. 1 People's Hospital, Yancheng, Jiangsu Province, China
| | - Xing-Hui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Pan Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Zhang-Chun Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Tian-Mei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
70
|
Zhang R, Zhang Y, Yi S. Identification of critical growth factors for peripheral nerve regeneration. RSC Adv 2019; 9:10760-10765. [PMID: 35515307 PMCID: PMC9062509 DOI: 10.1039/c9ra01710k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/26/2019] [Indexed: 12/17/2022] Open
Abstract
Growth factors are essential for the repair and regeneration of tissues and organs, including injured peripheral nerves. However, the expression changes of growth factors during peripheral nerve regeneration have not been fully elucidated. To obtain a global view of alternations of growth factors during the regeneration process, we explored previously achieved sequencing data of rat sciatic nerve stumps at 0 h, 1 d, 4 d, 7 d, and 14 d after nerve crush injury and screened differentially expressed upstream growth factors using Ingenuity Pathway Analysis (IPA) bioinformatic software. Differentially expressed growth factors were then subjected to Gene Ontology (GO) annotation and Kyoto Enrichment of Genes and Genomes (KEGG) pathway analysis. Regulatory networks of the differentially expressed growth factors in axon growth-related biological processes were constructed. Pivotal growth factors involved in axon growth were identified and validated by qRT-PCR. Our current study identified differentially expressed growth factors in the injured nerve stumps after peripheral nerve injury, discovered key growth factors for axon growth and nerve regeneration, and might facilitate the discovery of potential therapeutic targets of peripheral nerve injury. Growth factors are essential for the repair and regeneration of tissues and organs, including injured peripheral nerves.![]()
Collapse
Affiliation(s)
- Ruirui Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education
- Co-innovation Center of Neuroregeneration
- Nantong University
- Nantong
- China
| | - Yan Zhang
- Department of Respiratory and Critical Care Medicine
- Affiliated Hospital of Nantong University
- Nantong
- China
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education
- Co-innovation Center of Neuroregeneration
- Nantong University
- Nantong
- China
| |
Collapse
|
71
|
Nerve grafting for peripheral nerve injuries with extended defect sizes. Wien Med Wochenschr 2018; 169:240-251. [PMID: 30547373 PMCID: PMC6538587 DOI: 10.1007/s10354-018-0675-6] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 11/21/2018] [Indexed: 12/25/2022]
Abstract
Artificial and non-artificial nerve grafts are the gold standard in peripheral nerve reconstruction in cases with extensive loss of nerve tissue, particularly where a direct end-to-end suture or an autologous nerve graft is inauspicious. Different materials are marketed and approved by the US Food and Drug Administration (FDA) for peripheral nerve graft reconstruction. The most frequently used materials are collagen and poly(DL-lactide-ε-caprolactone). Only one human nerve allograft is listed for peripheral nerve reconstruction by the FDA. All marketed nerve grafts are able to demonstrate sufficient nerve regeneration over small distances not exceeding 3.0 cm. A key question in the field is whether nerve reconstruction on large defect lengths extending 4.0 cm or more is possible. This review gives a summary of current clinical and experimental approaches in peripheral nerve surgery using artificial and non-artificial nerve grafts in short and long distance nerve defects. Strategies to extend nerve graft lengths for long nerve defects, such as enhancing axonal regeneration, include the additional application of Schwann cells, mesenchymal stem cells or supporting co-factors like growth factors on defect sizes between 4.0 and 8.0 cm.
Collapse
|
72
|
Lo Furno D, Mannino G, Pellitteri R, Zappalà A, Parenti R, Gili E, Vancheri C, Giuffrida R. Conditioned Media From Glial Cells Promote a Neural-Like Connexin Expression in Human Adipose-Derived Mesenchymal Stem Cells. Front Physiol 2018; 9:1742. [PMID: 30555356 PMCID: PMC6282092 DOI: 10.3389/fphys.2018.01742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
The expression of neuronal and glial connexins (Cxs) has been evaluated in adipose-derived mesenchymal stem cells (ASCs) whose neural differentiation was promoted by a conditioned medium (CM) obtained from cultures of olfactory ensheathing cells (OECs) or Schwann cells (SCs). By immunocytochemistry and flow cytometer analysis it was found that Cx43 was already considerably expressed in naïve ASCs and further increased after 24 h and 7 days from CM exposition. Cx32 and Cx36 were significantly improved in conditioned cultures compared to control ASCs, whereas a decreased expression was noticed in the absence of CM treatments. Cx47 was virtually absent in any conditions. Altogether, high basal levels and induced increases of Cx43 expression suggest a potential attitude of ASCs toward an astrocyte differentiation, whereas the lack of Cx47 would indicate a poor propensity of ASCs to become oligodendrocytes. CM-evoked Cx32 and Cx36 increases showed that a neuronal- or a SC-like differentiation can be promoted by using this strategy. Results further confirm that environmental cues can favor an ASC neural differentiation, either as neuronal or glial elements. Of note, the use of glial products present in CM rather than the addition of chemical agents to achieve such differentiation would resemble "more physiological" conditions of differentiation. As a conclusion, the overexpression of typical neural Cxs would indicate the potential capability of neural-like ASCs to interact with neighboring neural cells and microenvironment.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosalia Pellitteri
- Institute of Neurological Sciences, National Research Council, Catania, Italy
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Elisa Gili
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| |
Collapse
|
73
|
Farzamfar S, Ehterami A, Salehi M, Vaeez A, Atashi A, Sahrapeyma H. Unrestricted Somatic Stem Cells Loaded in Nanofibrous Conduit as Potential Candidate for Sciatic Nerve Regeneration. J Mol Neurosci 2018; 67:48-61. [DOI: 10.1007/s12031-018-1209-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/08/2018] [Indexed: 12/25/2022]
|
74
|
Labroo P, Hilgart D, Davis B, Lambert C, Sant H, Gale B, Shea JE, Agarwal J. Drug-delivering nerve conduit improves regeneration in a critical-sized gap. Biotechnol Bioeng 2018; 116:143-154. [PMID: 30229866 DOI: 10.1002/bit.26837] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/03/2023]
Abstract
Autologous nerve grafts are the current "gold standard" for repairing large nerve gaps. However, they cause morbidity at the donor nerve site and only a limited amount of nerve can be harvested. Nerve conduits are a promising alternative to autografts and can act as guidance cues for the regenerating axons, without the need to harvest donor nerve. Separately, it has been shown that localized delivery of GDNF can enhance axon growth and motor recovery. FK506, an FDA approved small molecule, has also been shown to enhance peripheral nerve regeneration. This paper describes the design of a novel hole-based drug delivery apparatus integrated with a polytetrafluoroethylene (PTFE) nerve conduit for controlled local delivery of a protein such as GDNF or a small molecule such as FK506. The PTFE devices were tested in a diffusion chamber, and the bioactivity of the released media was evaluated by measuring neurite growth of dorsal root ganglions (DRGs) exposed to the released drugs. The drug delivering nerve guide was able to release bioactive concentrations of FK506 or GDNF. Following these tests, optimized drug releasing nerve conduits were implanted across 10 mm sciatic nerve gaps in a BL6 yellow fluorescent protein (YFP) mouse model, where they demonstrated significant improvement in muscle mass, compound muscle action potential, and axon myelination in vivo as compared with nerve conduits without the drug. The drug delivery nerve guide could release drug for extended periods of time and enhance axon growth in vitro and in vivo.
Collapse
Affiliation(s)
- Pratima Labroo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - David Hilgart
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Brett Davis
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Christopher Lambert
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Himanshu Sant
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Bruce Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Jill E Shea
- Department of Surgery, University of Utah, Salt Lake City, Utah
| | - Jayant Agarwal
- Department of Surgery, University of Utah, Salt Lake City, Utah
| |
Collapse
|
75
|
Li X, Wang X, Wang X, Chen H, Zhang X, Zhou L, Xu T. 3D bioprinted rat Schwann cell-laden structures with shape flexibility and enhanced nerve growth factor expression. 3 Biotech 2018; 8:342. [PMID: 30073127 PMCID: PMC6063810 DOI: 10.1007/s13205-018-1341-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/06/2018] [Indexed: 02/08/2023] Open
Abstract
Three-dimensional (3D) bioprinting composite alginate-gelatin hydrogel has encouraged the fabrication of cell-laden functional structures with cells from various tissues. However, reports focusing on printing this hydrogel for nerve tissue research are limited. This study aims at building in vitro Schwann cell 3D microenvironment with customized shapes through 3D bioprinting technology. Rat Schwann cell RSC96s encapsulated in composite alginate-gelatin hydrogel were printed with an extrusion-based bioprinter. Cells maintained high viability of 85.35 ± 6.19% immediately after printing and the printed hydrogel supported long-term Schwann cell proliferation for 2 weeks. Furthermore, after 14 days of culturing, Schwann cells cultured in printed structures maintained viability of 92.34 ± 2.19% and showed enhanced capability of nerve growth factor (NGF) release (142.41 ± 8.99 pg/ml) compared with cells from two-dimensional culture (92.27 ± 9.30 pg/ml). Specific Schwann cell marker S100β was also expressed by cells in printed structures. These printed structures may have the potential to be used as in vitro neurotrophic factor carriers and could be integrated into complex biomimetic artificial structures with the assistance of 3D bioprinting technology.
Collapse
Affiliation(s)
- Xinda Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084 People’s Republic of China
| | - Xiong Wang
- Biomanufacturing Engineering Research Laboratory, Graduate School at Shenzhen Tsinghua University, Shenzhen, 518055 People’s Republic of China
| | - Xuanzhi Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004 People’s Republic of China
| | - Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, 710032 People’s Republic of China
| | - Xinzhi Zhang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084 People’s Republic of China
- Medprin Biotech GmbH, Gutleutstr 163-167, 60327 Frankfurt, Germany
| | - Lian Zhou
- Department of Stomatology, Peking Union Medical College Hospital, CAMS and PUMC, Beijing, 100730 People’s Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084 People’s Republic of China
- Biomanufacturing Engineering Research Laboratory, Graduate School at Shenzhen Tsinghua University, Shenzhen, 518055 People’s Republic of China
- Department of Precision Medicine and Healthcare, Tsinghua Berkeley Shenzhen Institute, Shenzhen, 518055 People’s Republic of China
| |
Collapse
|
76
|
Zhang D, Wu S, Feng J, Duan Y, Xing D, Gao C. Micropatterned biodegradable polyesters clicked with CQAASIKVAV promote cell alignment, directional migration, and neurite outgrowth. Acta Biomater 2018; 74:143-155. [PMID: 29768188 DOI: 10.1016/j.actbio.2018.05.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/28/2018] [Accepted: 05/11/2018] [Indexed: 12/31/2022]
Abstract
The interplay of microstructures and biological cues is critical to regulate the behaviors of Schwann cells (SCs) in terms of cellular spatial arrangement and directional migration as well as neurite orientation for bridging the proximal and distal stumps of the injured peripheral nervous system. In this study, stripe micropatterns having ridges/grooves of width 20/20 and 20/40 μm were fabricated on the surface of maleimide-functionalized biodegradable poly(ester carbonate) (P(LLA-MTMC)) films by the polydimethylsiloxane mold-pressing method, respectively. The laminin-derived CQAASIKVAV peptides end-capped with an SH group were then grafted by the thiol-ene click reaction under mild conditions to obtain micropatterned and peptide-grafted films. SCs cultured on these films, especially on the 20/40-μm film, displayed faster and aligned adhesion as well as a larger number of elongated cells with a higher length-to-width (L/W) ratio along the stripe direction than those on the flat-pep film. The migration rate of SCs was significantly enhanced in parallel to the stripe direction with a large net displacement. The micropatterned and peptide-grafted films, especially the 20/40-μm film, could promote SC proliferation and nerve growth factor (NGF) secretion in a manner similar to that of the peptide-grafted planar film. Moreover, the neurites of rat pheochromocytoma 12 (PC12) cells sprouted along the ridges with a longer average length on the micropatterned and peptide-grafted films. The synergistic effect of physical patterns and biological cues was evaluated by considering the results of cell adhesion force; immunofluorescence staining of vinculin; fluorescence staining of F-actin and the nucleus; as well as gene expression of neural cadherin (NCAD), neurocan (NCAN), and myelin protein zero (P0). STATEMENT OF SIGNIFICANCE The interplay of microstructures and biological cues is critical to regulate the behaviors of Schwann cells (SCs) and nerve cells, and thereby the regeneration of peripheral nerve system. In this study, the combined micropatterning and CQAASIKVAV grafting endowed the modified P(LLA-MTMC) films with both contact guidance and bioactive chemical cues to enhance cell proliferation, directional alignment and migration, longer net displacement and larger NGF secretion, and stronger neurite outgrowth of SCs and PC12 cells. Hence, the integration of physical micropatterns and bioactive molecules is an effective way to obtain featured biomaterials for the regeneration of nerves and other types of tissues.
Collapse
|
77
|
Li Y, Sun Y, Cai M, Zhang H, Gao N, Huang H, Cui S, Yao D. Fas Ligand Gene (Faslg) Plays an Important Role in Nerve Degeneration and Regeneration After Rat Sciatic Nerve Injury. Front Mol Neurosci 2018; 11:210. [PMID: 29970988 PMCID: PMC6018423 DOI: 10.3389/fnmol.2018.00210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/28/2018] [Indexed: 01/09/2023] Open
Abstract
Wallerian degeneration (WD) is associated with changes in the expression levels of a large number of genes. However, the effects of these up- or down-regulated genes are poorly understood. We have reported some key factors that are differentially regulated during WD in our previous research. Here, we explored the roles of Fas ligand gene (Faslg) in WD after rat sciatic nerve injury. The data showed that Faslg was up-regulated in injured nerves. Expression changed of Faslg in Schwann cells (SCs) resulted in alterations in the release of related factors. Silencing or overexpression of Faslg affected SC proliferation, migration, and apoptosis through β-catenin, nuclear factor-κB (NF-κB), and caspase-3 pathways in vivo and in vitro. Our data suggest that Faslg is a key regulatory gene that affects nerve repair and regeneration in peripheral nerve injury. This study sheds new light on the effects of Faslg on peripheral nerve degeneration and/or regeneration.
Collapse
Affiliation(s)
- Yuting Li
- School of Life Sciences, Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Yuhua Sun
- School of Life Sciences, Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China.,Genetic Laboratory, Lianyungang Maternal and Child Health Hospital, Lianyungang, China
| | - Min Cai
- School of Life Sciences, Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Huanhuan Zhang
- School of Life Sciences, Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Nannan Gao
- School of Life Sciences, Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Huiwei Huang
- School of Medicine, Nantong University, Nantong, China
| | - Shusen Cui
- Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Dengbing Yao
- School of Life Sciences, Co-innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
78
|
Wang J, Tian L, He L, Chen N, Ramakrishna S, So KF, Mo X. Lycium barbarum polysaccharide encapsulated Poly lactic-co-glycolic acid Nanofibers: cost effective herbal medicine for potential application in peripheral nerve tissue engineering. Sci Rep 2018; 8:8669. [PMID: 29875468 PMCID: PMC5989206 DOI: 10.1038/s41598-018-26837-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 05/14/2018] [Indexed: 11/09/2022] Open
Abstract
Nerve regeneration is a serious clinical challenge following peripheral nerve injury. Lycium barbarum polysaccharide (LBP) is the major component of wolfberry extract, which has been shown to be neuroprotective and promising in nerve recovery in many studies. Electrospun nanofibers, especially core-shell structured nanofibers being capable of serving as both drug delivery system and tissue engineering scaffolds, are well known to be suitable scaffolds for regeneration of peripheral nerve applications. In this study, LBP was incorporated into core-shell structured nanofibrous scaffolds via coaxial electrospinning. Alamar blue assays were performed to investigate the proliferation of both PC12 and Schwann cells cultured on the scaffolds. The neuronal differentiation of PC12 cells was evaluated by NF200 expression with immunostaining and morphology changes observed by SEM. The results indicated that the released LBP dramatically enhanced both proliferation and neuronal differentiation of PC12 cells induced by NGF. Additionally, the promotion of Schwann cells myelination and neurite outgrowth of DRG neurons were also observed on LBP loaded scaffolds by LSCM with immunostaining. In summary, LBP, as a drug with neuroprotection, encapsulated into electrospun nanofibers could be a potential candidate as tissue engineered scaffold for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
- Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore, 117576, Singapore
| | - Lingling Tian
- Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore, 117576, Singapore
| | - Liumin He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Nuan Chen
- Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore, 117576, Singapore
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore, 117576, Singapore
- Guangdong-Hongkong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, 510632, China.
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China.
| | - Xiumei Mo
- State Key Laboratory of Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
79
|
Ko KR, Lee J, Lee D, Nho B, Kim S. Hepatocyte Growth Factor (HGF) Promotes Peripheral Nerve Regeneration by Activating Repair Schwann Cells. Sci Rep 2018; 8:8316. [PMID: 29844434 PMCID: PMC5973939 DOI: 10.1038/s41598-018-26704-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/17/2018] [Indexed: 01/12/2023] Open
Abstract
During the peripheral nerve regeneration process, a variety of neurotrophic factors play roles in nerve repair by acting on neuronal or non-neuronal cells. In this report, we investigated the role(s) of hepatocyte growth factor (HGF) and its receptor, c-met, in peripheral nerve regeneration. When mice were subjected to sciatic nerve injury, the HGF protein level was highly increased at the injured and distal sites. The level of both total and phosphorylated c-met was also highly upregulated, but almost exclusively in Schwann cells (SCs) distal from the injury site. When mice were treated with a c-met inhibitor, PHA-665752, myelin thickness and axon regrowth were decreased indicating that re-myelination was hindered. HGF promoted the migration and proliferation of cultured SCs, and also induced the expression of various genes such as GDNF and LIF, presumably by activating ERK pathways. Furthermore, exogenous supply of HGF around the injury site, by intramuscular injection of a plasmid DNA expressing human HGF, enhanced the myelin thickness and axon diameter in injured nerves. Taken together, our results indicate that HGF and c-met play important roles in Schwann cell-mediated nerve repair, and also that HGF gene transfer may provide a useful tool for treating peripheral neuropathy.
Collapse
Affiliation(s)
- Kyeong Ryang Ko
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea.,Viro Med, Co., Ltd, Seoul, 08826, Korea
| | | | | | - Boram Nho
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea
| | - Sunyoung Kim
- School of Biological Sciences, Seoul National University, Seoul, 08826, Korea. .,Viro Med, Co., Ltd, Seoul, 08826, Korea.
| |
Collapse
|
80
|
Wu S, Chen MS, Maurel P, Lee YS, Bunge MB, Arinzeh TL. Aligned fibrous PVDF-TrFE scaffolds with Schwann cells support neurite extension and myelination in vitro. J Neural Eng 2018; 15:056010. [PMID: 29794323 DOI: 10.1088/1741-2552/aac77f] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Polyvinylidene fluoride-trifluoroethylene (PVDF-TrFE), which is a piezoelectric, biocompatible polymer, holds promise as a scaffold in combination with Schwann cells (SCs) for spinal cord repair. Piezoelectric materials can generate electrical activity in response to mechanical deformation, which could potentially stimulate spinal cord axon regeneration. Our goal in this study was to investigate PVDF-TrFE scaffolds consisting of aligned fibers in supporting SC growth and SC-supported neurite extension and myelination in vitro. APPROACH Aligned fibers of PVDF-TrFE were fabricated using the electrospinning technique. SCs and dorsal root ganglion (DRG) explants were co-cultured to evaluate SC-supported neurite extension and myelination on PVDF-TrFE scaffolds. MAIN RESULTS PVDF-TrFE scaffolds supported SC growth and neurite extension, which was further enhanced by coating the scaffolds with Matrigel. SCs were oriented and neurites extended along the length of the aligned fibers. SCs in co-culture with DRGs on PVDF-TrFE scaffolds promoted longer neurite extension as compared to scaffolds without SCs. In addition to promoting neurite extension, SCs also formed myelin around DRG neurites on PVDF-TrFE scaffolds. SIGNIFICANCE This study demonstrated PVDF-TrFE scaffolds containing aligned fibers supported SC-neurite extension and myelination. The combination of SCs and PVDF-TrFE scaffolds may be a promising tissue engineering strategy for spinal cord repair.
Collapse
Affiliation(s)
- Siliang Wu
- Materials Science and Engineering Program, New Jersey Institute of Technology, Newark, NJ 07102, United States of America
| | | | | | | | | | | |
Collapse
|
81
|
Wang J, Tian L, Luo B, Ramakrishna S, Kai D, Loh XJ, Yang IH, Deen GR, Mo X. Engineering PCL/lignin nanofibers as an antioxidant scaffold for the growth of neuron and Schwann cell. Colloids Surf B Biointerfaces 2018; 169:356-365. [PMID: 29803151 DOI: 10.1016/j.colsurfb.2018.05.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/28/2018] [Accepted: 05/09/2018] [Indexed: 01/30/2023]
Abstract
Antioxidant is critical for the successful of nerve tissue regeneration, and biomaterials with antioxidant activity might be favorable for peripheral nerve repair. Lignin, a biopolymer from wood with excellent antioxidant properties, is still "unexplored" as biomaterials. To design an antioxidative bioscaffold for nerve regeneration, here we synthesized lignin-polycaprolactone (PCL) copolymers via solvent free ring-opening polymerization (ROP). Then such lignin-PCL copolymers were incorporated with PCL and engineered into nanofibrous scaffolds for supporting the growth of neuron and Schwann cell. Our results showed that the addition of lignin-PCL enhanced the mechanical properties of PCL nanofibers and endowed them with good antioxidant properties (up to 98.3 ± 1.9% free radical inhibition within 4 h). Cell proliferation assay showed that PCL/lignin-PCL nanofibers increased cell viability compared to PCL fibers, especially after an oxidative challenge. Moreover, Schwann cells and dorsal root ganglion (DRG) neurons cultured on the nanofibers to assess their potential for nerve regeneration. These results suggested that nanofibers with lignin copolymers promoted cell proliferation of both BMSCs and Schwann cells, enhanced myelin basic protein expressions of Schwann cells and stimulated neurite outgrowth of DRG neurons. In all, these sustainable, intrinsically antioxidant nanofibers may be a potential candidate for nerve TE applications.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, 117576, Singapore
| | - Lingling Tian
- Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, 117576, Singapore
| | - Baiwen Luo
- Singapore Institute for Neurotechnology, National University of Singapore, 28 Medical Drive, #05-COR, 119077, Singapore
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology, E3-05-14, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, 2 Engineering Drive 3, 117576, Singapore
| | - Dan Kai
- Institute of Materials Research and Engineering (IMRE), A*STAR, 2 Fusionopolis Way. Innovis, #08-03, 138634, Singapore.
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), A*STAR, 2 Fusionopolis Way. Innovis, #08-03, 138634, Singapore; Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, 117576, Singapore; Singapore Eye Research Institute, 11 Third Hospital Avenue, 168751, Singapore
| | - In Hong Yang
- Singapore Institute for Neurotechnology, National University of Singapore, 28 Medical Drive, #05-COR, 119077, Singapore
| | - G Roshan Deen
- Soft Materials Laboratory, Natural Sciences and Science Education, National Institute of Education Nanyang Technological University, Singapore
| | - Xiumei Mo
- State Key Laboratory of Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| |
Collapse
|
82
|
Lo Furno D, Mannino G, Giuffrida R, Gili E, Vancheri C, Tarico MS, Perrotta RE, Pellitteri R. Neural differentiation of human adipose-derived mesenchymal stem cells induced by glial cell conditioned media. J Cell Physiol 2018; 233:7091-7100. [PMID: 29737535 DOI: 10.1002/jcp.26632] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/30/2018] [Indexed: 12/15/2022]
Abstract
Adipose-derived mesenchymal stem cells (ASCs) may transdifferentiate into cells belonging to mesodermal, endodermal, and ectodermal lineages. The aim of this study was to verify whether a neural differentiation of ASCs could be induced by a conditioned medium (CM) obtained from cultures of olfactory ensheathing cells (OECs) or Schwann cells (SCs). ASCs were isolated from the stromal vascular fraction of adipose tissue and expanded for 2-3 passages. They were then cultured in OEC-CM or SC-CM for 24 hr or 7 days. At each stage, the cells were tested by immunocytochemistry and flow cytometer analysis to evaluate the expression of typical neural markers such as Nestin, PGP 9.5, MAP2, Synapsin I, and GFAP. Results show that both conditioned media induced similar positive effects, as all tested markers were overexpressed, especially at day 7. Overall, an evident trend toward neuronal or glial differentiation was not clearly detectable in many cases. Nevertheless, a higher tendency toward a neuronal phenotype was recognized for OEC-CM (considering MAP2 increases). On the other hand, SC-CM would be responsible for a more marked glial induction (considering GFAP increases). These findings confirm that environmental features can induce ASCs toward a neural differentiation, either as neuronal or glial elements. Rather than supplementing the culture medium by adding chemical agents, a "more physiological" condition was obtained here by means of soluble factors (cytokines/growth factors) likely released by glial cells. This culture strategy may provide valuable information in the development of cell-based therapeutic approaches for pathologies affecting the central/peripheral nervous system.
Collapse
Affiliation(s)
- Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Giuliana Mannino
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania, Italy
| | - Elisa Gili
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria S Tarico
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Rosario E Perrotta
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Rosalia Pellitteri
- Institute of Neurological Sciences, National Research Council, Section of Catania, Catania, Italy
| |
Collapse
|
83
|
Zhang R, Rosen JM. The role of undifferentiated adipose-derived stem cells in peripheral nerve repair. Neural Regen Res 2018; 13:757-763. [PMID: 29862994 PMCID: PMC5998619 DOI: 10.4103/1673-5374.232457] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
Peripheral nerve injuries impose significant health and economic consequences, yet no surgical repair can deliver a complete recovery of sensory or motor function. Traditional methods of repair are less than ideal: direct coaptation can only be performed when tension-free repair is possible, and transplantation of nerve autograft can cause donor-site morbidity and neuroma formation. Cell-based therapy delivered via nerve conduits has thus been explored as an alternative method of nerve repair in recent years. Stem cells are promising sources of the regenerative core material in a nerve conduit because stem cells are multipotent in function, abundant in supply, and more accessible than the myelinating Schwann cells. Among different types of stem cells, undifferentiated adipose-derived stem cell (uASC), which can be processed from adipose tissue in less than two hours, is a promising yet underexplored cell type. Studies of uASC have emerged in the past decade and have shown that autologous uASCs are non-immunogenic, easy to access, abundant in supply, and efficacious at promoting nerve regeneration. Two theories have been proposed as the primary regenerative mechanisms of uASC: in situ trans-differentiation towards Schwann cells, and secretion of trophic and anti-inflammatory factors. Future studies need to fully elucidate the mechanisms, side effects, and efficacy of uASC-based nerve regeneration so that uASCs can be utilized in clinical settings.
Collapse
Affiliation(s)
- Rui Zhang
- Dartmouth Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Joseph M. Rosen
- Dartmouth Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
84
|
Wu Y, Wang L, Hu T, Ma PX, Guo B. Conductive micropatterned polyurethane films as tissue engineering scaffolds for Schwann cells and PC12 cells. J Colloid Interface Sci 2018; 518:252-262. [DOI: 10.1016/j.jcis.2018.02.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/10/2018] [Accepted: 02/12/2018] [Indexed: 12/13/2022]
|
85
|
Up-Regulation of Cdc37 Contributes to Schwann Cell Proliferation and Migration After Sciatic Nerve Crush. Neurochem Res 2018; 43:1182-1190. [PMID: 29687307 DOI: 10.1007/s11064-018-2535-6] [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] [Received: 11/08/2017] [Revised: 04/02/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022]
Abstract
Cell division cycle protein 37 (Cdc37), a molecular chaperone takes part in a series of cellular processes including cell signal transduction, cell cycle progression, cell proliferation, cell motility, oncogenesis and malignant progression. It can not only recruit immature protein kinases to HSP90 but also work alone. Cdc37 was reported to be associated with neurogenesis, neurite outgrowth, axon guidance and myelination. However, the roles of Cdc37 on Schwann cells (SC) after peripheral nerve injury (PNI) remain unknown. In this study, we found that the expression of Cdc37 increased and reached the peak at 1 week after sciatic nerve crush (SNC), which was consistent with that of proliferation cell nuclear antigen. Immunofluorescence verified that Cdc37 co-localized with SC in vivo and in vitro. Intriguingly, Cdc37 protein level was potentiated in the model of TNF-α-induced SC proliferation. Moreover, we found that Cdc37 silencing impaired proliferation of SC in vitro. Moreover, Cdc37 suppression attenuated kinase signaling pathways of Raf-ERK and PI3K/AKT which are crucial cell signaling for SC proliferation. Finally, we found that Cdc37 silencing inhibited SC migration in vitro. In conclusion, we demonstrated that the way Cdc37 contributed to SC proliferation is likely via activating kinase signaling pathways of Raf-ERK and PI3K/AKT, and CDC37 was also involved in SC migration after SNC.
Collapse
|
86
|
Abstract
The schwann cells of the peripheral nervous system are indispensable for the formation, maintenance, and modulation of synapses over the life cycle. They not only recognize neuron-glia signaling molecules, but also secrete gliotransmitters. Through these processes, they regulate neuronal excitability and thus the release of neurotransmitters from the nerve terminal at the neuromuscular junction. Gliotransmitters strongly affect nerve communication, and their secretion is mainly triggered by synchronized Ca2+ signaling, implicating Ca2+ waves in synapse function. Reciprocally, neurotransmitters released during synaptic activity can evoke increases in intracellular Ca2+ levels. A reconsideration of the interplay between the two main types of cells in the nervous system is due, as the concept of nervous system activity comprising only neuron-neuron and neuron-muscle action has become untenable. A more precise understanding of the roles of schwann cells in nerve-muscle signaling is required.
Collapse
Affiliation(s)
- Sujin Hyung
- Department of Mechanical and Aerospace Engineering, Seoul National University, San 56-1, Shilim-dong, Gwanak-gu, Seoul, 08826, Korea
- BK21 Plus Transformative Training Program for Creative Mechanical and Aerospace Engineers, Seoul National University, San 56-1, Shilim-dong, Gwanak-gu, Seoul, 08826, Korea
| | - Kyuhwan Jung
- Graduate Program of Nano Science and Technology, Graduate School of Yonsei University, 50-1 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Sung-Rae Cho
- Graduate Program of Nano Science and Technology, Graduate School of Yonsei University, 50-1 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Korea
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 50-1 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Noo Li Jeon
- Department of Mechanical and Aerospace Engineering, Seoul National University, San 56-1, Shilim-dong, Gwanak-gu, Seoul, 08826, Korea
| |
Collapse
|
87
|
AAV1.NT-3 gene therapy increases muscle fiber diameter through activation of mTOR pathway and metabolic remodeling in a CMT mouse model. Gene Ther 2018. [PMID: 29523879 DOI: 10.1038/s41434-018-0009-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurotrophin 3 (NT-3) has well-recognized effects on peripheral nerve and Schwann cells, promoting axonal regeneration and associated myelination. In this study, we assessed the effects of AAV.NT-3 gene therapy on the oxidative state of the neurogenic muscle from the TremblerJ (Tr J ) mice at 16 weeks post-gene injection and found that the muscle fiber size increase was associated with a change in the oxidative state of muscle fibers towards normalization of the fiber type ratio seen in the wild type. NT-3-induced fiber size increase was most prominent for the fast twitch glycolytic fiber population. These changes in the Tr J muscle were accompanied by increased phosphorylation levels of 4E-BP1 and S6 proteins as evidence of mTORC1 activation. In parallel, the expression levels of the mitochondrial biogenesis regulator PGC1α, and the markers of glycolysis (HK1 and PK1) increased in the TrJ muscle. In vitro studies showed that recombinant NT-3 can directly induce Akt/mTOR pathway activation in the TrkC expressing myotubes but not in myoblasts. In addition, myogenin expression levels were increased in myotubes while p75 NTR expression was downregulated compared to myoblasts, indicating that NT-3 induced myoblast differentiation is associated with mTORC1 activation. These studies for the first time have shown that NT-3 increases muscle fiber diameter in the neurogenic muscle through direct activation of mTOR pathway and that the fiber size increase is more prominent for fast twitch glycolytic fibers.
Collapse
|
88
|
Ward PJ, Clanton SL, English AW. Optogenetically enhanced axon regeneration: motor versus sensory neuron-specific stimulation. Eur J Neurosci 2018; 47:294-304. [PMID: 29363200 DOI: 10.1111/ejn.13836] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 01/05/2018] [Accepted: 01/11/2018] [Indexed: 12/25/2022]
Abstract
Brief neuronal activation in injured peripheral nerves is both necessary and sufficient to enhance motor axon regeneration, and this effect is specific to the activated motoneurons. It is less clear whether sensory neurons respond in a similar manner to neuronal activation following peripheral axotomy. Further, it is unknown to what extent enhancement of axon regeneration with increased neuronal activity relies on a reflexive interaction within the spinal circuitry. We used mouse genetics and optical tools to evaluate the precision and selectivity of system-specific neuronal activation to enhance axon regeneration in a mixed nerve. We evaluated sensory and motor axon regeneration in two different mouse models expressing the light-sensitive cation channel, channelrhodopsin (ChR2). We selectively activated either sensory or motor axons using light stimulation combined with transection and repair of the sciatic nerve. Regardless of genotype, the number of ChR2-positive neurons whose axons had regenerated successfully was greater following system-specific optical treatment, with no effect on the number of ChR2-negative neurons (whether motor or sensory neurons). We conclude that acute system-specific neuronal activation is sufficient to enhance both motor and sensory axon regeneration. This regeneration-enhancing effect is likely cell autonomous.
Collapse
Affiliation(s)
- Patricia J Ward
- Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Room 425, Atlanta, GA, 30322, USA
| | - Scott L Clanton
- Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Room 425, Atlanta, GA, 30322, USA
| | - Arthur W English
- Department of Cell Biology, Emory University School of Medicine, 615 Michael Street, Room 425, Atlanta, GA, 30322, USA
| |
Collapse
|
89
|
D’Arpa S, Zabbia G, Cannizzaro C, Salimbeni G, Plescia F, Mariolo AV, Cassata G, Cicero L, Puleio R, Martorana A, Moschella F, Cordova A. Seeding nerve sutures with minced nerve-graft (MINE-G): a simple method to improve nerve regeneration in rats. Acta Chir Belg 2018; 118:27-35. [PMID: 28738725 DOI: 10.1080/00015458.2017.1353237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The aim of this study was to assess the effect of seeding the distal nerve suture with nerve fragments in rats. METHODS On 20 rats, a 15 mm sciatic nerve defect was reconstructed with a nerve autograft. In the Study Group (10 rats), a minced 1 mm nerve segment was seeded around the nerve suture. In the Control Group (10 rats), a nerve graft alone was used. At 4 and 12 weeks, a walking track analysis with open field test (WTA), hystomorphometry (number of myelinated fibers (n), fiber density (FD) and fiber area (FA) and soleus and gastrocnemius muscle weight ratios (MWR) were evaluated. The Student t-test was used for statistical analysis. RESULTS At 4 and 12 weeks the Study Group had a significantly higher n and FD (p = .043 and .033). The SMWR was significantly higher in the Study Group at 12 weeks (p = .0207). CONCLUSIONS Seeding the distal nerve suture with nerve fragments increases the number of myelinated fibers, the FD and the SMWR. The technique seems promising and deserves further investigation to clarify the mechanisms involved and its functional effects.
Collapse
Affiliation(s)
- Salvatore D’Arpa
- Plastische Heelkunde, Universitair Ziekenhuis Gent, Gent, Belgium
| | - Giovanni Zabbia
- Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Carla Cannizzaro
- Department of Sciences for Health Promotion and Mother and Child Care ‘GIUSEPPE D’ALESSANDRO’, University of Palermo, Palermo, Italy
| | | | - Fulvio Plescia
- Department of Sciences for Health Promotion and Mother and Child Care ‘GIUSEPPE D’ALESSANDRO’, University of Palermo, Palermo, Italy
| | - Alessio Vincenzo Mariolo
- Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Giovanni Cassata
- Laboratory Animal House/Unit, Institute of Experimental Zooprophylactic of Sicily, Palermo, Italy
| | - Luca Cicero
- Laboratory Animal House/Unit, Institute of Experimental Zooprophylactic of Sicily, Palermo, Italy
| | - Roberto Puleio
- Histopathology and Immunohistochemistry Laboratory, Institute Experimental Zooprophylactic of Sicily, Palermo, Italy
| | - Anna Martorana
- Department of Human Pathology, University of Palermo, Palermo, Italy
| | - Francesco Moschella
- Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Adriana Cordova
- Division of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| |
Collapse
|
90
|
Novel miR-sc4 regulates the proliferation and migration of Schwann cells by targeting Cdk5r1. Mol Cell Biochem 2018; 447:209-215. [DOI: 10.1007/s11010-018-3305-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/25/2018] [Indexed: 12/22/2022]
|
91
|
Cervellini I, Galino J, Zhu N, Allen S, Birchmeier C, Bennett DL. Sustained MAPK/ERK Activation in Adult Schwann Cells Impairs Nerve Repair. J Neurosci 2018; 38:679-690. [PMID: 29217688 PMCID: PMC5777114 DOI: 10.1523/jneurosci.2255-17.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/29/2017] [Accepted: 10/19/2017] [Indexed: 11/21/2022] Open
Abstract
The MAPK/ERK pathway has a critical role in PNS development. It is required for Schwann cell (SC) differentiation and myelination; sustained embryonic MAPK/ERK activation in SCs enhances myelin growth overcoming signals that normally end myelination. Excess activation of this pathway can be maladaptive as in adulthood acute strong activation of MAPK/ERK has been shown to cause SC dedifferentiation and demyelination. We used a mouse model (including male and female animals) in which the gain-of-function MEK1DD allele produces sustained MAPK/ERK activation in adult SCs, and we determined the impact of such activation on nerve repair. In the uninjured nerve, MAPK/ERK activation neither impaired myelin nor reactivated myelination. However, in the injured nerve it was detrimental and resulted in delayed repair and functional recovery. In the early phase of injury, the rate of myelin clearance was faster. Four weeks following injury, when nerve repair is normally advanced, myelinated axons of MEK1DD mutants demonstrated higher rates of myelin decompaction, a reduced number of Cajal bands. and decreased internodal length. We noted the presence of abnormal Remak bundles with long SCs processes and reduced numbers of C-fibers/Remak bundle. Both the total number of regenerating axons and the intraepidermal nerve fiber density in the skin were reduced. Sustained activation of MAPK/ERK in adult SCs is therefore deleterious to successful nerve repair, emphasizing the differences in the signaling processes coordinating nerve development and repair. Our results also underline the key role of SCs in axon regeneration and successful target reinnervation.SIGNIFICANCE STATEMENT The MAPK/ERK pathway promotes developmental myelination and its sustained activation in SCs induced continuous myelin growth, compensating for the absence of essential myelination signals. However, the strength of activation is fundamental because acute strong induction of MAPK/ERK in adulthood induces demyelination. What has been unknown is the effect of a mild but sustained MAPK/ERK activation in SCs on nerve repair in adulthood. This promoted myelin clearance but led to abnormalities in nonmyelinating and myelinating SCs in the later phases of nerve repair, resulting in slowed axon regeneration, cutaneous reinnervation, and functional recovery. Our results emphasize the distinct role of the MAPK/ERK pathway in developmental myelination versus remyelination and the importance of signaling between SCs and axons for successful axon regeneration.
Collapse
Affiliation(s)
- Ilaria Cervellini
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Jorge Galino
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Ning Zhu
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Shannen Allen
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction Group, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - David L Bennett
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| |
Collapse
|
92
|
Glial differentiation of human inferior turbinate-derived stem cells: a new source of cells for nerve repair. Neuroreport 2018; 28:235-241. [PMID: 28169963 DOI: 10.1097/wnr.0000000000000731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Schwann cell (SC) transplantation as a cell-based therapy can enhance peripheral and central nerve repair experimentally, but it is limited by donor site morbidity for clinical application. We investigated whether human turbinate-derived mesenchymal stem cells (hTMSCs) isolated from discarded inferior turbinate during surgery can differentiate into functional SC-like cells. hTMSCs expressed mesenchymal cell surface markers CD29, CD44, CD90, and CD105 and did not express neural crest markers P75 and Nestin. After monolayer culture in predifferentiation medium and transdifferentiation medium with a mixture of glial growth factors and chemical regents for 14 days, the differentiated hTMSCs exhibited a spindle-like morphology similar to that of SCs. RT-PCR, immunocytochemical staining, and western blotting analysis indicated that SC-like cells expressed the glial markers S100β, P75, and glial fibrillary acidic protein at the gene and protein level. Compared with hTMSCs, differentiated hTMSCs secreted more neurotrophins, and significantly enhanced the neurite length when cocultured with dorsal root ganglia neuronal cells. Our data indicated that hTMSCs can differentiate into functional SC-like cells and have the ability to facilitate the neurite growth of dorsal root ganglia neuronal cells in vitro, representing a promising source of cells for nerve repair.
Collapse
|
93
|
Aydin T, Gurcan C, Taheri H, Yilmazer A. Graphene Based Materials in Neural Tissue Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:129-142. [PMID: 29882208 DOI: 10.1007/5584_2018_221] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to its extraordinary features such as large surface area, high electrical conductivity, chemical stability and mechanical properties, graphene attracts great interest in various fields of biomedical sciences including biosensors, cancer therapy, diagnosis and regenerative medicine. The use of graphene-based materials has been of great interest for the design of scaffolds that can promote neural tissue regeneration. Recent studies published over the last few years clearly show that graphene and graphene based materials promote adhesion, proliferation and differentiation of various cells including embryonic stem cells (ESC), neural stem cells (NSC), mesenchymal stem cells (MSC) and induced pluripotent stem cells (iPSC). Therefore graphene based materials are one of the promising nanoplatforms in regenerative medicine for neural tissue injury. With its unique topographic and chemical properties, graphene is used as a scaffold that could provide a bridge between regenerating nerves. More importantly, as a conductive substrate, graphene allows the continuation of electrical conduction between damaged nerve ends. The integration of supportive cells such as glial, neural precursor or stem cells in such a scaffold shows higher regeneration when compared to currently used neural autografts and nerve conduits. This review discusses the details of such studies involving graphene based materials with a special interest on neural stem cells, mesenchymal stem cells or pluripotent stem cells.
Collapse
Affiliation(s)
- Tugce Aydin
- Biotechnology Institute, Ankara University, Tandogan/Ankara, Turkey.,Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey
| | - Cansu Gurcan
- Biotechnology Institute, Ankara University, Tandogan/Ankara, Turkey.,Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey
| | - Hadiseh Taheri
- Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey
| | - Açelya Yilmazer
- Engineering Faculty, Biomedical Engineering Department, Ankara University, Tandogan/Ankara, Turkey. .,Stem Cell Institute, Ankara University, Balgat/Ankara, Turkey.
| |
Collapse
|
94
|
Brick RM, Sun AX, Tuan RS. Neurotrophically Induced Mesenchymal Progenitor Cells Derived from Induced Pluripotent Stem Cells Enhance Neuritogenesis via Neurotrophin and Cytokine Production. Stem Cells Transl Med 2017; 7:45-58. [PMID: 29215199 PMCID: PMC5746147 DOI: 10.1002/sctm.17-0108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022] Open
Abstract
Adult tissue‐derived mesenchymal stem cells (MSCs) are known to produce a number of bioactive factors, including neurotrophic growth factors, capable of supporting and improving nerve regeneration. However, with a finite culture expansion capacity, MSCs are inherently limited in their lifespan and use. We examined here the potential utility of an alternative, mesenchymal‐like cell source, derived from induced pluripotent stem cells, termed induced mesenchymal progenitor cells (MiMPCs). We found that several genes were upregulated and proteins were produced in MiMPCs that matched those previously reported for MSCs. Like MSCs, the MiMPCs secreted various neurotrophic and neuroprotective factors, including brain‐derived neurotrophic factor (BDNF), interleukin‐6 (IL‐6), leukemia inhibitory factor (LIF), osteopontin, and osteonectin, and promoted neurite outgrowth in chick embryonic dorsal root ganglia (DRG) cultures compared with control cultures. Cotreatment with a pharmacological Trk‐receptor inhibitor did not result in significant decrease in MiMPC‐induced neurite outgrowth, which was however inhibited upon Jak/STAT3 blockade. These findings suggest that the MiMPC induction of DRG neurite outgrowth is unlikely to be solely dependent on BDNF, but instead Jak/STAT3 activation by IL‐6 and/or LIF is likely to be critical neurotrophic signaling pathways of the MiMPC secretome. Taken together, these findings suggest MiMPCs as a renewable, candidate source of therapeutic cells and a potential alternative to MSCs for peripheral nerve repair, in view of their ability to promote nerve growth by producing many of the same growth factors and cytokines as Schwann cells and signaling through critical neurotrophic pathways. stemcellstranslational Medicine2018;7:45–58
Collapse
Affiliation(s)
- Rachel M Brick
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Aaron X Sun
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
95
|
Masciullo C, Dell'Anna R, Tonazzini I, Böettger R, Pepponi G, Cecchini M. Hierarchical thermoplastic rippled nanostructures regulate Schwann cell adhesion, morphology and spatial organization. NANOSCALE 2017; 9:14861-14874. [PMID: 28948996 DOI: 10.1039/c7nr02822a] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Periodic ripples are a variety of anisotropic nanostructures that can be realized by ion beam irradiation on a wide range of solid surfaces. Only a few authors have investigated these surfaces for tuning the response of biological systems, probably because it is challenging to directly produce them in materials that well sustain long-term cellular cultures. Here, hierarchical rippled nanotopographies with a lateral periodicity of ∼300 nm are produced from a gold-irradiated germanium mold in polyethylene terephthalate (PET), a biocompatible polymer approved by the US Food and Drug Administration for clinical applications, by a novel three-step embossing process. The effects of nano-ripples on Schwann Cells (SCs) are studied in view of their possible use for nerve-repair applications. The data demonstrate that nano-ripples can enhance short-term SC adhesion and proliferation (3-24 h after seeding), drive their actin cytoskeleton spatial organization and sustain long-term cell growth. Notably, SCs are oriented perpendicularly with respect to the nanopattern lines. These results provide information about the possible use of hierarchical nano-rippled elements for nerve-regeneration protocols.
Collapse
Affiliation(s)
- Cecilia Masciullo
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, 56127 Pisa, Italy.
| | | | | | | | | | | |
Collapse
|
96
|
Naseri-Nosar M, Salehi M, Hojjati-Emami S. Cellulose acetate/poly lactic acid coaxial wet-electrospun scaffold containing citalopram-loaded gelatin nanocarriers for neural tissue engineering applications. Int J Biol Macromol 2017; 103:701-708. [DOI: 10.1016/j.ijbiomac.2017.05.054] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/30/2017] [Accepted: 05/13/2017] [Indexed: 12/22/2022]
|
97
|
Assis AD, de Assis Araújo F, Dos Santos RAS, Andrade SP, Zanon RG. Pattern of Mas expression in acute and post-acute stage of nerve injury in mice. Peptides 2017; 96:15-19. [PMID: 28870798 DOI: 10.1016/j.peptides.2017.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/12/2017] [Accepted: 08/28/2017] [Indexed: 12/24/2022]
Abstract
Angiotensin-(1-7) (Ang [1-7]) and its receptor Mas are involved in a number of physiological processes, including control of arterial pressure and modulation of nervous system actions. However, the involvement of the Ang-(1-7)/Mas axis in peripheral nerve injury has not been investigated. Using a model of sciatic nerve injury in mice, we demonstrated opposing changes in Mas receptor expression at days 2 and 14 post-injury. Mas receptor expression was more intense 2days after the nerve lesion, compared with the intensity of the intact nerve. At this time point, the sciatic nerve functional index was -20. At day 14 after the lesion, the intensity of the immunostaining labeling in longitudinal sections of the nerve was reduced (∼30%) and the functional index increased +36 (gait improvement). In the axotomized group treated with A779 (a Mas receptor antagonist), the functional recovery index decreased in relation to the untreated axotomized group. The Mas receptor inhibitor also altered the intensity of labeling of S-100, GAP43, and IBA-1 (morphological features compatible with delayed axon growth). This study demonstrated that Ang-(1-7)/Mas axis activity was differentially modulated in the acute and post-acute stages of nerve injury.
Collapse
Affiliation(s)
- Alex Dias Assis
- Department of Human Anatomy, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil
| | - Fernanda de Assis Araújo
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil
| | | | - Silvia Passos Andrade
- Department of Physiology, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Renata Graciele Zanon
- Department of Human Anatomy, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil.
| |
Collapse
|
98
|
Bierlein De la Rosa M, Sharma AD, Mallapragada SK, Sakaguchi DS. Transdifferentiation of brain-derived neurotrophic factor (BDNF)-secreting mesenchymal stem cells significantly enhance BDNF secretion and Schwann cell marker proteins. J Biosci Bioeng 2017; 124:572-582. [PMID: 28694020 DOI: 10.1016/j.jbiosc.2017.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/09/2017] [Accepted: 05/23/2017] [Indexed: 01/03/2023]
Abstract
The use of genetically modified mesenchymal stem cells (MSCs) is a rapidly growing area of research targeting delivery of therapeutic factors for neuro-repair. Cells can be programmed to hypersecrete various growth/trophic factors such as brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and nerve growth factor (NGF) to promote regenerative neurite outgrowth. In addition to genetic modifications, MSCs can be subjected to transdifferentiation protocols to generate neural cell types to physically and biologically support nerve regeneration. In this study, we have taken a novel approach by combining these two unique strategies and evaluated the impact of transdifferentiating genetically modified MSCs into a Schwann cell-like phenotype. After 8 days in transdifferentiation media, approximately 30-50% of transdifferentiated BDNF-secreting cells immunolabeled for Schwann cell markers such as S100β, S100, and p75NTR. An enhancement was observed 20 days after inducing transdifferentiation with minimal decreases in expression levels. BDNF production was quantified by ELISA, and its biological activity tested via the PC12-TrkB cell assay. Importantly, the bioactivity of secreted BDNF was verified by the increased neurite outgrowth of PC12-TrkB cells. These findings demonstrate that not only is BDNF actively secreted by the transdifferentiated BDNF-MSCs, but also that it has the capacity to promote neurite sprouting and regeneration. Given the fact that BDNF production remained stable for over 20 days, we believe that these cells have the capacity to produce sustainable, effective, BDNF concentrations over prolonged time periods and should be tested within an in vivo system for future experiments.
Collapse
Affiliation(s)
- Metzere Bierlein De la Rosa
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Anup D Sharma
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA; Neuroscience Program, Iowa State University, Ames, IA 50011, USA
| | - Surya K Mallapragada
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA; Neuroscience Program, Iowa State University, Ames, IA 50011, USA
| | - Donald S Sakaguchi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA; Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA; Neuroscience Program, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
99
|
Schaal SM, Kitay BM, Cho KS, Lo TP, Barakat DJ, Marcillo AE, Sanchez AR, Andrade CM, Pearse DD. Schwann Cell Transplantation Improves Reticulospinal Axon Growth and Forelimb Strength after Severe Cervical Spinal Cord Contusion. Cell Transplant 2017; 16:207-28. [PMID: 17503734 DOI: 10.3727/000000007783464768] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Schwann cell (SC) implantation alone has been shown to promote the growth of propriospinal and sensory axons, but not long-tract descending axons, after thoracic spinal cord injury (SCI). In the current study, we examined if an axotomy close to the cell body of origin (so as to enhance the intrinsic growth response) could permit supraspinal axons to grow onto SC grafts. Adult female Fischer rats received a severe (C5) cervical contusion (1.1 mm displacement, 3 KDyn). At 1 week postinjury, 2 million SCs ex vivo transduced with lentiviral vector encoding enhanced green fluorescent protein (EGFP) were implanted within media into the injury epicenter; injury-only animals served as controls. Animals were tested weekly using the BBB score for 7 weeks postimplantation and received at end point tests for upper body strength: self-supported forelimb hanging, forearm grip force, and the incline plane. Following behavioral assessment, animals were anterogradely traced bilaterally from the reticular formation using BDA-Texas Red. Stereological quantification revealed a twofold increase in the numbers of preserved NeuN+ neurons rostral and caudal to the injury/graft site in SC implanted animals, corroborating previous reports of their neuroprotective efficacy. Examination of labeled reticulospinal axon growth revealed that while rarely an axon was present within the lesion site of injury-only controls, numerous reticulospinal axons had penetrated the SC implant/lesion milieu. This has not been observed following implantation of SCs alone into the injured thoracic spinal cord. Significant behavioral improvements over injury-only controls in upper limb strength, including an enhanced grip strength (a 296% increase) and an increased self-supported forelimb hanging, accompanied SC-mediated neuroprotection and reticulospinal axon growth. The current study further supports the neuroprotective efficacy of SC implants after SCI and demonstrates that SCs alone are capable of supporting modest supraspinal axon growth when the site of axon injury is closer to the cell body of the axotomized neuron.
Collapse
Affiliation(s)
- S M Schaal
- The Miami Project to Cure Paralysis, University of Miami School of Medicine, Miami, FL 33101, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Acosta MC, Copley PA, Harrell JR, Wilhelm JC. Estrogen signaling is necessary for exercise-mediated enhancement of motoneuron participation in axon regeneration after peripheral nerve injury in mice. Dev Neurobiol 2017; 77:1133-1143. [PMID: 28388831 DOI: 10.1002/dneu.22501] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 11/09/2022]
Abstract
Thousands of people each year suffer from peripheral nerve injury. Treatment options are limited, and recovery is often incomplete. Treadmill exercise can enhance nerve regeneration; however, this appears to occur in a sex-dependent manner. Females respond best to short duration, high speed interval training; whereas, males respond best to slower, continuous training. Previous studies have shown a role for testosterone in this process, but the role of estrogen is unknown. To evaluate the role of estrogen signaling in treadmill exercise, we blocked estrogen receptor (ER) signaling during treadmill exercise in males and female wild type mice. The right common fibular (CF) branch of the sciatic nerve was cut and repaired with fibrin glue that contained the ER antagonist ICI 182,780. Estradiol-filled or blank Silastic capsules were implanted subcutaneously at the time of nerve transection. Starting three days post-transection, exercised mice received treadmill training using the paradigm appropriate to their sex 5 days a week for 2 weeks. Fourteen days after the initial nerve transection, motoneurons whose axons had regenerated at least 1.5 mm distal to the original cut sites were labeled with a retrograde tracer. Regeneration was quantified by counting the number of fluorescent labeled motoneurons in the lumbar region of the spinal cord. Both treadmill training and estradiol administration increased the number of motoneurons participating in axon regeneration, but these effects were blocked by ER antagonist treatment. Estrogen signaling is important for the enhancing effects of treadmill exercise on motoneuron participation after peripheral nerve cut. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1133-1143, 2017.
Collapse
Affiliation(s)
- Melina C Acosta
- Department of Psychology, College of Charleston, Charleston, South Carolina
| | - Patricia A Copley
- Department of Psychology, College of Charleston, Charleston, South Carolina
| | - Jamie R Harrell
- Department of Psychology, College of Charleston, Charleston, South Carolina
| | - Jennifer C Wilhelm
- Department of Psychology, College of Charleston, Charleston, South Carolina
| |
Collapse
|