1
|
Suzuki T, Kadoya K, Endo T, Yamasaki M, Watanabe M, Iwasaki N. GFRα1 Promotes Axon Regeneration after Peripheral Nerve Injury by Functioning as a Ligand. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2400812. [PMID: 39630029 PMCID: PMC11775530 DOI: 10.1002/advs.202400812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 11/04/2024] [Indexed: 01/30/2025]
Abstract
The neurotrophic factor, Glial cell line derived neurotrophi factor (GDNF), exerts a variety of biological effects through binding to its receptors, GDNF family receptor alpha-1 (GFRα1), and RET. However, the existence of cells expressing GFRα1 but not RET raises the possibility that GFRα1 can function independently from RET. Here, it is shown that GFRα1 released from repair Schwann cells (RSCs) functions as a ligand in a GDNF-RET-independent manner to promote axon regeneration after peripheral nerve injury (PNI). Local administration of GFRα1 into injured nerve promoted axon regeneration, even more when combined with GDNF blockade. GFRα1 bound to a receptor complex consisting of NCAM and integrin α7β1 of dorsal root ganglion neurons in a GDNF-RET independent manner. This is further confirmed by the Ret Y1062F knock-in mice, which cannot transmit most of GDNF-RET signaling. Finally, local administration of GFRα1 into injured sciatic nerve promoted functional recovery. These findings reveal a novel role of GFRα1 as a ligand, the molecular mechanism supporting axon regeneration by RSCs, and a novel therapy for peripheral nerve repair.
Collapse
Affiliation(s)
- Tomoaki Suzuki
- Department of Orthopaedic SurgeryGraduate School of MedicineHokkaido UniversitySapporoHokkaido0608638Japan
| | - Ken Kadoya
- Department of Orthopaedic SurgeryGraduate School of MedicineHokkaido UniversitySapporoHokkaido0608638Japan
| | - Takeshi Endo
- Department of Orthopaedic SurgeryGraduate School of MedicineHokkaido UniversitySapporoHokkaido0608638Japan
| | - Miwako Yamasaki
- Department of AnatomyGraduate School of Medicine, Hokkaido UniversitySapporoHokkaido0608638Japan
| | - Masahiko Watanabe
- Department of AnatomyGraduate School of Medicine, Hokkaido UniversitySapporoHokkaido0608638Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic SurgeryGraduate School of MedicineHokkaido UniversitySapporoHokkaido0608638Japan
| |
Collapse
|
2
|
Jiang S, Chen B, Sun ZY. Enhancing therapeutic potential: Human adipose-derived mesenchymal stem cells modified with recombinant adeno-associated virus expressing VEGF165 gene for peripheral nerve injury. Kaohsiung J Med Sci 2024; 40:819-829. [PMID: 39101328 DOI: 10.1002/kjm2.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/26/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
This study aimed to investigate the therapeutic potential of human adipose-derived mesenchymal stem cells (hADSCs) modified with recombinant adeno-associated virus (rAAV) carrying the vascular endothelial growth factor 165 (VEGF165) gene in peripheral nerve injury (PNI). The hADSCs were categorized into blank, control (transduced with rAAV control vector), and VEGF165 (transduced with rAAV VEGF165 vector) groups. Subsequently, Schwann cell differentiation was induced, and Schwann cell markers were assessed. The sciatic nerve injury mouse model received injections of phosphate-buffered saline (PBS group), PBS containing hADSCs (hADSCs group), rAAV control vector (control-hADSCs group), or rAAV VEGF165 vector (VEGF165-hADSCs group) into the nerve defect site. Motor function recovery, evaluated through the sciatic function index (SFI), and nerve regeneration, assessed via toluidine blue staining along with scrutiny of Schwann cell markers and neurotrophic factors, were conducted. Modified hADSCs exhibited enhanced Schwann cell differentiation and elevated expression of Schwann cell markers [S100 calcium-binding protein B (S100B), NGF receptor (NGFR), and glial fibrillary acidic protein (GFAP)]. Mice in the VEGF165-hADSCs group demonstrated improved motor function recovery compared to those in the other three groups, accompanied by increased fiber diameter, axon diameter, and myelin thickness, as well as elevated expression of Schwann cell markers (S100B, NGFR, and GFAP) and neurotrophic factors [mature brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF)] in the distal nerve segment. rAAV-VEGF165 modification enhances hADSC potential in PNI, promoting motor recovery and nerve regeneration. Elevated Schwann cell markers and neurotrophic factors underscore therapy benefits, providing insights for nerve injury strategies.
Collapse
Affiliation(s)
- Shuai Jiang
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Bo Chen
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Zhen-Yu Sun
- Department of Orthopedics, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| |
Collapse
|
3
|
Bengur FB, Komatsu C, Fedor CN, Loder S, Baker JS, Totwani A, Irgebay Z, Nerone WV, Solari MG, Marra KG. Biodegradable Nerve Guide with Glial Cell Line-Derived Neurotrophic Factor Improves Recovery After Facial Nerve Injury in Rats. Facial Plast Surg Aesthet Med 2023; 25:478-486. [PMID: 36877591 PMCID: PMC10664574 DOI: 10.1089/fpsam.2022.0346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Background: Bioengineered nerve guides with glial cell line-derived neurotrophic factor (GDNF) support recovery after facial nerve injury by acting as regenerative scaffolds. Objective: To compare functional, electrophysiological, and histological outcomes after repair of rat facial nerve transection in control, empty nerve guide, and nerve guide with GDNF conditions. Methods: Rats underwent transection and primary repair of the buccal branch of the facial nerve and were divided into (1) transection and repair only, (2) transection and repair augmented with empty guide, (3) transection and repair augmented with GDNF-guide groups. Weekly measurements of the whisking movements were recorded. At 12 weeks, compound muscle action potentials (CMAPs) at the whisker pad were assessed, and samples were collected for histomorphometric analysis. Results: Rats in GDNF-guide group displayed the earliest peak in normalized whisking amplitude. CMAPs were significantly higher after GDNF-guide placement. Mean fiber surface area of the target muscle, axonal count of the injured branch, and the number of Schwann cells were highest with GDNF guides. Conclusion: The biodegradable nerve guide containing double-walled GDNF microspheres enhanced recovery after facial nerve transection and primary repair.
Collapse
Affiliation(s)
- Fuat Baris Bengur
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chiaki Komatsu
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Caroline Nadia Fedor
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shawn Loder
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jocelyn S. Baker
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aanchal Totwani
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhazira Irgebay
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - W. Vincent Nerone
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mario G. Solari
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kacey G. Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Zhang WJ, Liu SC, Ming LG, Yu JW, Zuo C, Hu DX, Luo HL, Zhang Q. Potential role of Schwann cells in neuropathic pain. Eur J Pharmacol 2023; 956:175955. [PMID: 37541365 DOI: 10.1016/j.ejphar.2023.175955] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/26/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Neuropathic pain (NPP) is a common syndrome associated with most forms of disease, which poses a serious threat to human health. NPP may persist even after the nociceptive stimulation is eliminated, and treatment is extremely challenging in such cases. Schwann cells (SCs) form the myelin sheaths around neuronal axons and play a crucial role in neural information transmission. SCs can secrete trophic factors to nourish and protect axons, and can further secrete pain-related factors to induce pain. SCs may be activated by peripheral nerve injury, triggering the transformation of myelinated and non-myelinated SCs into cell phenotypes that specifically promote repair. These differentiated SCs provide necessary signals and spatial clues for survival, axonal regeneration, and nerve regeneration of damaged neurons. They can further change the microenvironment around the regions of nerve injury, and relieve the pain by repairing the injured nerve. Herein, we provide a comprehensive overview of the biological characteristics of SCs, discuss the relationship between SCs and nerve injury, and explore the potential mechanism of SCs and the occurrence of NPP. Moreover, we summarize the feasible strategies of SCs in the treatment of NPP, and attempt to elucidate the deficiencies and defects of SCs in the treatment of NPP.
Collapse
Affiliation(s)
- Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Si-Cheng Liu
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Li-Guo Ming
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Jian-Wen Yu
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Cheng Zuo
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Hong-Liang Luo
- Department of Gastrointestinal surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China.
| | - Qiao Zhang
- Orthopedics Department, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China.
| |
Collapse
|
5
|
Chernov AV, Shubayev VI. Sexual dimorphism of early transcriptional reprogramming in degenerating peripheral nerves. Front Mol Neurosci 2022; 15:1029278. [DOI: 10.3389/fnmol.2022.1029278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Sexual dimorphism is a powerful yet understudied factor that influences the timing and efficiency of gene regulation in axonal injury and repair processes in the peripheral nervous system. Here, we identified common and distinct biological processes in female and male degenerating (distal) nerve stumps based on a snapshot of transcriptional reprogramming 24 h after axotomy reflecting the onset of early phase Wallerian degeneration (WD). Females exhibited transcriptional downregulation of a larger number of genes than males. RhoGDI, ERBB, and ERK5 signaling pathways increased activity in both sexes. Males upregulated genes and canonical pathways that exhibited robust baseline expression in females in both axotomized and sham nerves, including signaling pathways controlled by neuregulin and nerve growth factors. Cholesterol biosynthesis, reelin signaling, and synaptogenesis signaling pathways were downregulated in females. Signaling by Rho Family GTPases, cAMP-mediated signaling, and sulfated glycosaminoglycan biosynthesis were downregulated in both sexes. Estrogens potentially influenced sex-dependent injury response due to distinct regulation of estrogen receptor expression. A crosstalk of cytokines and growth hormones could promote sexually dimorphic transcriptional responses. We highlighted prospective regulatory activities due to protein phosphorylation, extracellular proteolysis, sex chromosome-specific expression, major urinary proteins (MUPs), and genes involved in thyroid hormone metabolism. Combined with our earlier findings in the corresponding dorsal root ganglia (DRG) and regenerating (proximal) nerve stumps, sex-specific and universal early phase molecular triggers of WD enrich our knowledge of transcriptional regulation in peripheral nerve injury and repair.
Collapse
|
6
|
Chernov AV, Shubayev VI. Sexually dimorphic transcriptional programs of early-phase response in regenerating peripheral nerves. Front Mol Neurosci 2022; 15:958568. [PMID: 35983069 PMCID: PMC9378824 DOI: 10.3389/fnmol.2022.958568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
The convergence of transcriptional and epigenetic changes in the peripheral nervous system (PNS) reshapes the spatiotemporal gene expression landscape in response to nerve transection. The control of these molecular programs exhibits sexually dimorphic characteristics that remain not sufficiently characterized. In the present study, we recorded genome-wide and sex-dependent early-phase transcriptional changes in regenerating (proximal) sciatic nerve 24 h after axotomy. Male nerves exhibited more extensive transcriptional changes with male-dominant upregulation of cytoskeletal binding and structural protein genes. Regulation of mRNAs encoding ion and ionotropic neurotransmitter channels displayed prominent sexual dimorphism consistent with sex-specific mRNA axonal transport in an early-phase regenerative response. Protein kinases and axonal transport genes showed sexually dimorphic regulation. Genes encoding components of synaptic vesicles were at high baseline expression in females and showed post-injury induction selectively in males. Predictive bioinformatic analyses established patterns of sexually dimorphic regulation of neurotrophic and immune genes, including activation of glial cell line-derived neurotrophic factor Gfra1 receptor and immune checkpoint cyclin D1 (Ccnd1) potentially linked to X-chromosome encoded tissue inhibitor of matrix metallo proteinases 1 (Timp1). Regulatory networks involving Olig1, Pou3f3/Oct6, Myrf, and Myt1l transcription factors were linked to sex-dependent reprogramming in regenerating nerves. Differential expression patterns of non-coding RNAs motivate a model of sexually dimorphic nerve regenerative responses to injury determined by epigenetic factors. Combined with our findings in the corresponding dorsal root ganglia (DRG), unique early-phase sex-specific molecular triggers could enrich the mechanistic understanding of peripheral neuropathies.
Collapse
Affiliation(s)
- Andrei V. Chernov
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, United States
- VA San Diego Healthcare System, San Diego, CA, United States
- *Correspondence: Andrei V. Chernov,
| | - Veronica I. Shubayev
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, United States
- VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
7
|
Rhymes ER, Tosolini AP, Fellows AD, Mahy W, McDonald NQ, Schiavo G. Bimodal regulation of axonal transport by the GDNF-RET signalling axis in healthy and diseased motor neurons. Cell Death Dis 2022; 13:584. [PMID: 35798698 PMCID: PMC9263112 DOI: 10.1038/s41419-022-05031-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/08/2023]
Abstract
Deficits in axonal transport are one of the earliest pathological outcomes in several models of amyotrophic lateral sclerosis (ALS), including SOD1G93A mice. Evidence suggests that rescuing these deficits prevents disease progression, stops denervation, and extends survival. Kinase inhibitors have been previously identified as transport enhancers, and are being investigated as potential therapies for ALS. For example, inhibitors of p38 mitogen-activated protein kinase and insulin growth factor receptor 1 have been shown to rescue axonal transport deficits in vivo in symptomatic SOD1G93A mice. In this work, we investigated the impact of RET, the tyrosine kinase receptor for glial cell line-derived neurotrophic factor (GDNF), as a modifier of axonal transport. We identified the fundamental interplay between RET signalling and axonal transport in both wild-type and SOD1G93A motor neurons in vitro. We demonstrated that blockade of RET signalling using pharmacological inhibitors and genetic knockdown enhances signalling endosome transport in wild-type motor neurons and uncovered a divergence in the response of primary motor neurons to GDNF compared with cell lines. Finally, we showed that inhibition of the GDNF-RET signalling axis rescues in vivo transport deficits in early symptomatic SOD1G93A mice, promoting RET as a potential therapeutic target in the treatment of ALS.
Collapse
Affiliation(s)
- Elena R Rhymes
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, UK
| | - Andrew P Tosolini
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, UK
| | | | - William Mahy
- Alzheimer's Research UK UCL Drug Discovery Institute, University College London, London, UK
| | - Neil Q McDonald
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, UK.
- UK Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
8
|
Jessen KR, Mirsky R. The Role of c-Jun and Autocrine Signaling Loops in the Control of Repair Schwann Cells and Regeneration. Front Cell Neurosci 2022; 15:820216. [PMID: 35221918 PMCID: PMC8863656 DOI: 10.3389/fncel.2021.820216] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
After nerve injury, both Schwann cells and neurons switch to pro-regenerative states. For Schwann cells, this involves reprogramming of myelin and Remak cells to repair Schwann cells that provide the signals and mechanisms needed for the survival of injured neurons, myelin clearance, axonal regeneration and target reinnervation. Because functional repair cells are essential for regeneration, it is unfortunate that their phenotype is not robust. Repair cell activation falters as animals get older and the repair phenotype fades during chronic denervation. These malfunctions are important reasons for the poor outcomes after nerve damage in humans. This review will discuss injury-induced Schwann cell reprogramming and the concept of the repair Schwann cell, and consider the molecular control of these cells with emphasis on c-Jun. This transcription factor is required for the generation of functional repair cells, and failure of c-Jun expression is implicated in repair cell failures in older animals and during chronic denervation. Elevating c-Jun expression in repair cells promotes regeneration, showing in principle that targeting repair cells is an effective way of improving nerve repair. In this context, we will outline the emerging evidence that repair cells are sustained by autocrine signaling loops, attractive targets for interventions aimed at promoting regeneration.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
9
|
Basu S, Choudhury IN, Nazareth L, Chacko A, Shelper T, Vial ML, Ekberg JAK, St John JA. In vitro modulation of Schwann cell behavior by VEGF and PDGF in an inflammatory environment. Sci Rep 2022; 12:662. [PMID: 35027585 PMCID: PMC8758747 DOI: 10.1038/s41598-021-04222-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Peripheral glial cell transplantation with Schwann cells (SCs) is a promising approach for treating spinal cord injury (SCI). However, improvements are needed and one avenue to enhance regenerative functional outcomes is to combine growth factors with cell transplantation. Vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) are neuroprotective, and a combination of these factors has improved outcomes in rat SCI models. Thus, transplantation of SCs combined with VEGF and PDGF may further improve regenerative outcomes. First, however, we must understand how the two factors modulate SCs. In this in vitro study, we show that an inflammatory environment decreased the rate of SC-mediated phagocytosis of myelin debris but the addition of VEGF and PDGF (alone and combined) improved phagocytosis. Cytokine expression by SCs in the inflammatory environment revealed that addition of PDGF led to significantly lower level of pro-inflammatory cytokine, TNF-α, but IL-6 and anti-inflammatory cytokines (TGF-β and IL-10), remained unaltered. Further, PDGF was able to decrease the expression of myelination associated gene Oct6 in the presence of inflammatory environment. Overall, these results suggest that the use of VEGF and/or PDGF combined with SC transplantation may be beneficial in SCI therapy.
Collapse
Affiliation(s)
- Souptik Basu
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Indra N Choudhury
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Lynn Nazareth
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Anu Chacko
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Todd Shelper
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Marie-Laure Vial
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jenny A K Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - James A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia. .,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|
10
|
Advanced approaches to regenerate spinal cord injury: The development of cell and tissue engineering therapy and combinational treatments. Biomed Pharmacother 2021; 146:112529. [PMID: 34906773 DOI: 10.1016/j.biopha.2021.112529] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) devastate event that is commonly caused by traumatic or non-traumatic events. The reinnervation of spinal cord axons is hampered through a myriad of devices counting on the damaged myelin, inflammation, glial scar, and defective inhibitory molecules. Unfortunately, an effective treatment to completely repair SCI and improve functional recovery has not been found. In this regard, strategies such as using cells, biomaterials, biomolecules, and drugs have been reported to be effective for SCI recovery. Furthermore, recent advances in combinatorial treatments, which address various aspects of SCI pathophysiology, provide optimistic outcomes for spinal cord regeneration. According to the global importance of SCI, the goal of this article review is to provide an overview of the pathophysiology of SCI, with an emphasis on the latest modes of intervention and current advanced approaches for the treatment of SCI, in conjunction with an assessment of combinatorial approaches in preclinical and clinical trials. So, this article can give scientists and clinicians' clues to help them better understand how to construct preclinical and clinical studies that could lead to a breakthrough in spinal cord regeneration.
Collapse
|
11
|
Ki SM, Jeong HS, Lee JE. Primary Cilia in Glial Cells: An Oasis in the Journey to Overcoming Neurodegenerative Diseases. Front Neurosci 2021; 15:736888. [PMID: 34658775 PMCID: PMC8514955 DOI: 10.3389/fnins.2021.736888] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022] Open
Abstract
Many neurodegenerative diseases have been associated with defects in primary cilia, which are cellular organelles involved in diverse cellular processes and homeostasis. Several types of glial cells in both the central and peripheral nervous systems not only support the development and function of neurons but also play significant roles in the mechanisms of neurological disease. Nevertheless, most studies have focused on investigating the role of primary cilia in neurons. Accordingly, the interest of recent studies has expanded to elucidate the role of primary cilia in glial cells. Correspondingly, several reports have added to the growing evidence that most glial cells have primary cilia and that impairment of cilia leads to neurodegenerative diseases. In this review, we aimed to understand the regulatory mechanisms of cilia formation and the disease-related functions of cilia, which are common or specific to each glial cell. Moreover, we have paid close attention to the signal transduction and pathological mechanisms mediated by glia cilia in representative neurodegenerative diseases. Finally, we expect that this field of research will clarify the mechanisms involved in the formation and function of glial cilia to provide novel insights and ideas for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Soo Mi Ki
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hui Su Jeong
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Ji Eun Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Samsung Medical Center, Samsung Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
12
|
Choi SJ, Park SY, Shin YH, Heo SH, Kim KH, Lee HI, Kim JK. Mesenchymal Stem Cells Derived from Wharton's Jelly Can Differentiate into Schwann Cell-Like Cells and Promote Peripheral Nerve Regeneration in Acellular Nerve Grafts. Tissue Eng Regen Med 2021; 18:467-478. [PMID: 33515168 DOI: 10.1007/s13770-020-00329-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/08/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Schwann cells (SCs) secrete neurotrophic factors and provide structural support and guidance during axonal regeneration. However, nearby nerves may be damaged to obtain primary SCs, and there is a lack of nervous tissue donors. We investigated the potential of Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) in differentiating into Schwann cell-like cells (WJ-SCLCs) as an alternative to SCs. We also examined whether implantation of WJ-SCLCs-laden acellular nerve grafts (ANGs) are effective in inducing functional recovery and nerve regeneration in an animal model of peripheral nerve injury. METHODS The differentiation of WJ-MSCs into WJ-SCLCs was determined by analyzing SC-specific markers. The secretion of neurotrophic factors was assessed by the Neuro Discovery antibody array. Neurite outgrowth and myelination of axons were found in a co-culture system involving motor neuron cell lines. The effects of ANGs on repairing sciatic nerves were evaluated using video gait angle test, isometric tetanic force analysis, and toluidine blue staining. RESULTS Compared with undifferentiated WJ-MSCs, WJ-SCLCs showed higher expression levels of SC-specific markers such as S100β, GFAP, KROX20, and NGFR. WJ-SCLCs also showed higher secreted amounts of brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and granulocyte-colony stimulating factor than did WJ-MSCs. WJ-SCLCs effectively promoted the outgrowth and myelination of neurites in motor neuron cells, and WJ-SCLCs laden ANGs significantly facilitated peripheral nerve regeneration in an animal model of sciatic nerve injury. CONCLUSION WJ-MSCs were readily differentiated into WJ-SCLCs, which effectively promoted the regeneration of peripheral nerves. Transplantation of WJ-SCLCs with ANGs might be useful for assisting peripheral nerve regeneration.
Collapse
Affiliation(s)
- Soon Jin Choi
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea
| | - Suk Young Park
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea
| | - Young Ho Shin
- Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic Road 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Seung-Ho Heo
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Kang-Hyun Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Hyo In Lee
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Jae Kwang Kim
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea. .,Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic Road 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
13
|
Eggers R, de Winter F, Tannemaat MR, Malessy MJA, Verhaagen J. GDNF Gene Therapy to Repair the Injured Peripheral Nerve. Front Bioeng Biotechnol 2020; 8:583184. [PMID: 33251197 PMCID: PMC7673415 DOI: 10.3389/fbioe.2020.583184] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
A spinal root avulsion is the most severe proximal peripheral nerve lesion possible. Avulsion of ventral root filaments disconnects spinal motoneurons from their target muscles, resulting in complete paralysis. In patients that undergo brachial plexus nerve repair, axonal regeneration is a slow process. It takes months or even years to bridge the distance from the lesion site to the distal targets located in the forearm. Following ventral root avulsion, without additional pharmacological or surgical treatments, progressive death of motoneurons occurs within 2 weeks (Koliatsos et al., 1994). Reimplantation of the avulsed ventral root or peripheral nerve graft can act as a conduit for regenerating axons and increases motoneuron survival (Chai et al., 2000). However, this beneficial effect is transient. Combined with protracted and poor long-distance axonal regeneration, this results in permanent function loss. To overcome motoneuron death and improve functional recovery, several promising intervention strategies are being developed. Here, we focus on GDNF gene-therapy. We first introduce the experimental ventral root avulsion model and discuss its value as a proxy to study clinical neurotmetic nerve lesions. Second, we discuss our recent studies showing that GDNF gene-therapy is a powerful strategy to promote long-term motoneuron survival and improve function when target muscle reinnervation occurs within a critical post-lesion period. Based upon these observations, we discuss the influence of timing of the intervention, and of the duration, concentration and location of GDNF delivery on functional outcome. Finally, we provide a perspective on future research directions to realize functional recovery using gene therapy.
Collapse
Affiliation(s)
- Ruben Eggers
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Fred de Winter
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Martijn R Tannemaat
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Martijn J A Malessy
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Neurosurgery, Leiden University Medical Center, Leiden, Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Academy of Arts and Sciences, Amsterdam, Netherlands.,Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognition Research, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
RET-independent signaling by GDNF ligands and GFRα receptors. Cell Tissue Res 2020; 382:71-82. [PMID: 32737575 PMCID: PMC7529620 DOI: 10.1007/s00441-020-03261-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022]
Abstract
The discovery in the late 1990s of the partnership between the RET receptor tyrosine kinase and the GFRα family of GPI-anchored co-receptors as mediators of the effects of GDNF family ligands galvanized the field of neurotrophic factors, firmly establishing a new molecular framework besides the ubiquitous neurotrophins. Soon after, however, it was realized that many neurons and brain areas expressed GFRα receptors without expressing RET. These observations led to the formulation of two new concepts in GDNF family signaling, namely, the non-cell-autonomous functions of GFRα molecules, so-called trans signaling, as well as cell-autonomous functions mediated by signaling receptors distinct from RET, which became known as RET-independent signaling. To date, the best studied RET-independent signaling pathway for GDNF family ligands involves the neural cell adhesion molecule NCAM and its association with GFRα co-receptors. Among the many functions attributed to this signaling system are neuronal migration, neurite outgrowth, dendrite branching, spine formation, and synaptogenesis. This review summarizes our current understanding of this and other mechanisms of RET-independent signaling by GDNF family ligands and GFRα receptors, as well as their physiological importance.
Collapse
|
15
|
Zhang Y, Jiang K, Xie G, Ding J, Peng S, Liu X, Sun C, Tang X. FGF21 impedes peripheral myelin development by stimulating p38 MAPK/c-Jun axis. J Cell Physiol 2020; 236:1345-1361. [PMID: 32657446 DOI: 10.1002/jcp.29942] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/18/2020] [Accepted: 07/02/2020] [Indexed: 01/13/2023]
Abstract
Fibroblast growth factor 21 (FGF21) as a metabolic stress hormone, is mainly secreted by the liver. In addition to its well-defined roles in energy homeostasis, FGF21 has been shown to promote remyelination after injury in the central nervous system. In the current study, we sought to examine the potential roles of FGF21 in the peripheral nervous system (PNS) myelination. In the PNS myelin development, Fgf21 expression was reversely correlated with myelin gene expression. In cultured primary Schwann cells (SCs), the application of recombinant FGF21 greatly attenuates myelination-associated gene expression, including Oct6, Krox20, Mbp, Mpz, and Pmp22. Accordingly, the injection of FGF21 into neonatal rats markedly mitigates the myelination in sciatic nerves. On the contrary, the infusion of the anti-FGF21 antibody accelerates the myelination. Mechanistically, both extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) were stimulated by FGF21 in SCs and sciatic nerves. Following experiments including pharmaceutical intervention and gene manipulation revealed that the p38 MAPK/c-Jun axis, rather than ERK, is targeted by FGF21 for mediating its repression on myelination in SCs. Taken together, our data provide a new aspect of FGF21 by acting as a negative regulator for the myelin development process in the PNS via activation of p38 MAPK/c-Jun.
Collapse
Affiliation(s)
- Yunzhong Zhang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China.,School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Ketao Jiang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| | - Guoqing Xie
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| | - Jie Ding
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| | - Su Peng
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| | - Xiaoyu Liu
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| | - Cheng Sun
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| | - Xin Tang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregenetation, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
16
|
Qiu S, Rao Z, He F, Wang T, Xu Y, Du Z, Yao Z, Lin T, Yan L, Quan D, Zhu Q, Liu X. Decellularized nerve matrix hydrogel and glial-derived neurotrophic factor modifications assisted nerve repair with decellularized nerve matrix scaffolds. J Tissue Eng Regen Med 2020; 14:931-943. [PMID: 32336045 DOI: 10.1002/term.3050] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/11/2020] [Accepted: 04/20/2020] [Indexed: 12/28/2022]
Abstract
Nerve defects are challenging to address clinically without satisfactory treatments. As a reliable alternative to autografts, decellularized nerve matrix scaffolds (DNM-S) have been widely used in clinics for surgical nerve repair. However, DNM-S remain inferior to autografts in their ability to support nerve regeneration for long nerve defects. In this study, we systematically and clearly presented the nano-architecture of nerve-specific structures, including the endoneurium, basement membrane and perineurium/epineurium in DNM-S. Furthermore, we modified the DNM-S by supplementing decellularized nerve matrix hydrogel (DNMG) and glial-derived neurotrophic factor (GDNF) and then bridged a 50-mm sciatic nerve defect in a beagle model. Fifteen beagles were randomly divided into three groups (five per group): an autograft group, DNM-S group and GDNF-DNMG-modified DNM-S (DNM-S/GDNF@DNMG) group. DNM-S/GDNF@DNMG, as optimized nerve grafts, were used to bridge nerve defects in the same manner as in the DNM-S group. The repair outcome was evaluated by behavioural observations, electrophysiological assessments, regenerated nerve tissue histology and reinnervated target muscle examinations. Compared with the DNM-S group, limb function, electrophysiological responses and histological findings were improved in the DNM-S/GDNF@DNMG group 6 months after grafting, reflecting a narrower gap between the effects of DNM-S and autografts. In conclusion, modification of DNM-S with DNMG and GDNF enhanced nerve regeneration and functional recovery, indicating that noncellular modification of DNM-S is a promising method for treating long nerve defects.
Collapse
Affiliation(s)
- Shuai Qiu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilong Rao
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Fulin He
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Wang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yiwei Xu
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Zhaoyi Du
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Zhi Yao
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Lin
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liwei Yan
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Daping Quan
- Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Peripheral Nerve Tissue-Engineering and Technology Research Center, Guangzhou, China.,GD Functional Biomaterials Engineering Technology Research Center, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China.,PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Qingtang Zhu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Peripheral Nerve Tissue-Engineering and Technology Research Center, Guangzhou, China
| | - Xiaolin Liu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Peripheral Nerve Tissue-Engineering and Technology Research Center, Guangzhou, China
| |
Collapse
|
17
|
Wang A, Xiao Y, Huang P, Liu L, Xiong J, Li J, Mao D, Liu L. Novel NtA and LG1 Mutations in Agrin in a Single Patient Causes Congenital Myasthenic Syndrome. Front Neurol 2020; 11:239. [PMID: 32328026 PMCID: PMC7160337 DOI: 10.3389/fneur.2020.00239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/12/2020] [Indexed: 12/27/2022] Open
Abstract
Congenital myasthenic syndrome (CMS) is a group of genetic disorders of neuromuscular transmission that is characterized by muscle weakness. A mutation in the gene encoding agrin (AGRN) is a rare cause of CMS, and only a few families or isolated cases have been reported. We reported a pediatric proband exhibiting muscle weakness in the trunk and limbs with skeletal malformation and intellectual disability and performed whole-exome sequencing (WES) of the proband parent-offspring trio. Results revealed a new compound heterozygous mutation in AGRN: c.125A>C (p.Glu42Ala) in the N-terminal agrin domain (NtA) and c.4516G>A (p.Ala1506Thr) in the laminin G1 domain (LG1). Bioinformatic analysis predicted the mutation as possibly pathogenic. The new compound heterozygous mutation in AGRN may disrupt agrin's known function of bridging laminin and α-dystroglycan and undermine the formation and maintenance of the neuromuscular junction (NMJ) via both muscular and neural agrin pathways. It may also induce secondary peripheral neuropathy and skeletal malformation.
Collapse
Affiliation(s)
- Aiping Wang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Xiong
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jian Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ding'an Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatrics Neurology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Pan D, Mackinnon SE, Wood MD. Advances in the repair of segmental nerve injuries and trends in reconstruction. Muscle Nerve 2020; 61:726-739. [PMID: 31883129 DOI: 10.1002/mus.26797] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/18/2022]
Abstract
Despite advances in surgery, the reconstruction of segmental nerve injuries continues to pose challenges. In this review, current neurobiology regarding regeneration across a nerve defect is discussed in detail. Recent findings include the complex roles of nonneuronal cells in nerve defect regeneration, such as the role of the innate immune system in angiogenesis and how Schwann cells migrate within the defect. Clinically, the repair of nerve defects is still best served by using nerve autografts with the exception of small, noncritical sensory nerve defects, which can be repaired using autograft alternatives, such as processed or acellular nerve allografts. Given current clinical limits for when alternatives can be used, advanced solutions to repair nerve defects demonstrated in animals are highlighted. These highlights include alternatives designed with novel topology and materials, delivery of drugs specifically known to accelerate axon growth, and greater attention to the role of the immune system.
Collapse
Affiliation(s)
- Deng Pan
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Susan E Mackinnon
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew D Wood
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
19
|
Hwang K, Jung K, Kim IS, Kim M, Han J, Lim J, Shin JE, Jang JH, Park KI. Glial Cell Line-derived Neurotrophic Factor-overexpressing Human Neural Stem/Progenitor Cells Enhance Therapeutic Efficiency in Rat with Traumatic Spinal Cord Injury. Exp Neurobiol 2019; 28:679-696. [PMID: 31902156 PMCID: PMC6946112 DOI: 10.5607/en.2019.28.6.679] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) causes axonal damage and demyelination, neural cell death, and comprehensive tissue loss, resulting in devastating neurological dysfunction. Neural stem/progenitor cell (NSPCs) transplantation provides therapeutic benefits for neural repair in SCI, and glial cell linederived neurotrophic factor (GDNF) has been uncovered to have capability of stimulating axonal regeneration and remyelination after SCI. In this study, to evaluate whether GDNF would augment therapeutic effects of NSPCs for SCI, GDNF-encoding or mock adenoviral vector-transduced human NSPCs (GDNF-or Mock-hNSPCs) were transplanted into the injured thoracic spinal cords of rats at 7 days after SCI. Grafted GDNFhNSPCs showed robust engraftment, long-term survival, an extensive distribution, and increased differentiation into neurons and oligodendroglial cells. Compared with Mock-hNSPC- and vehicle-injected groups, transplantation of GDNF-hNSPCs significantly reduced lesion volume and glial scar formation, promoted neurite outgrowth, axonal regeneration and myelination, increased Schwann cell migration that contributed to the myelin repair, and improved locomotor recovery. In addition, tract tracing demonstrated that transplantation of GDNF-hNSPCs reduced significantly axonal dieback of the dorsal corticospinal tract (dCST), and increased the levels of dCST collaterals, propriospinal neurons (PSNs), and contacts between dCST collaterals and PSNs in the cervical enlargement over that of the controls. Finally grafted GDNF-hNSPCs substantially reversed the increased expression of voltage-gated sodium channels and neuropeptide Y, and elevated expression of GABA in the injured spinal cord, which are involved in the attenuation of neuropathic pain after SCI. These findings suggest that implantation of GDNF-hNSPCs enhances therapeutic efficiency of hNSPCs-based cell therapy for SCI.
Collapse
Affiliation(s)
- Kyujin Hwang
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea.,Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Kwangsoo Jung
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Il-Sun Kim
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Miri Kim
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jungho Han
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Joohee Lim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jeong Eun Shin
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Korea
| | - Kook In Park
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea.,Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea.,Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
20
|
A population of nonneuronal GFRα3-expressing cells in the bone marrow resembles nonmyelinating Schwann cells. Cell Tissue Res 2019; 378:441-456. [DOI: 10.1007/s00441-019-03068-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 07/01/2019] [Indexed: 12/17/2022]
|
21
|
Jonscher R, Belkind-Gerson J. Concise Review: Cellular and Molecular Mechanisms of Postnatal Injury-Induced Enteric Neurogenesis. Stem Cells 2019; 37:1136-1143. [PMID: 31145813 DOI: 10.1002/stem.3045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 05/14/2019] [Indexed: 12/20/2022]
Abstract
Although still controversial, there is increasing agreement that postnatal neurogenesis occurs in the enteric nervous system (ENS) in response to injury. Following acute colitis, there is significant cell death of enteric neurons and evidence suggests that subsequent neural regeneration follows. An enteric neural stem/progenitor cell population with neurogenic potential has been identified in culture; in vivo, compensatory neurogenesis is driven by enteric glia and may also include de-differentiated Schwann cells. Recent evidence suggests that changes in the enteric microenvironment due to injury-associated increases in glial cell-derived neurotrophic factor (GDNF), serotonin (5-hydroxytryptamine [HT]), products from the gut microbiome, and possibly endocannabinoids may lead to the transdifferentiation of mature enteric glia and may reprogram recruited Schwann cells. Targeting neurogenic pathways presents a promising avenue toward the development of new and innovative treatments for acquired damage to the ENS. In this review, we discuss potential sources of newly generated adult enteric neurons, the involvement of GDNF, 5-HT, endocannabinoids, and lipopolysaccharide, as well as therapeutic applications of this evolving work. Stem Cells 2019;37:1136-1143.
Collapse
Affiliation(s)
- Raleigh Jonscher
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jaime Belkind-Gerson
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA.,Neurogastroenterology Program, Digestive Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
22
|
The evolution and multi-molecular properties of NF1 cutaneous neurofibromas originating from C-fiber sensory endings and terminal Schwann cells at normal sites of sensory terminations in the skin. PLoS One 2019; 14:e0216527. [PMID: 31107888 PMCID: PMC6527217 DOI: 10.1371/journal.pone.0216527] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 04/24/2019] [Indexed: 12/30/2022] Open
Abstract
In addition to large plexiform neurofibromas (pNF), NF1 patients are frequently disfigured by cutaneous neurofibromas (cNF) and are often afflicted with chronic pain and itch even from seemingly normal skin areas. Both pNFs and cNF consist primarily of benign hyperproliferating nonmyelinating Schwann cells (nSC). While pNF clearly arise within deep nerves and plexuses, the role of cutaneous innervation in the origin of cNF and in chronic itch and pain is unknown. First, we conducted a comprehensive, multi-molecular, immunofluorescence (IF) analyses on 3mm punch biopsies from three separate locations in normal appearing, cNF-free skin in 19 NF1 patients and skin of 16 normal subjects. At least one biopsy in 17 NF1 patients had previously undescribed micro-lesions consisting of a small, dense cluster of nonpeptidergic C-fiber endings and the affiliated nSC consistently adjoining adnexal structures—dermal papillae, hair follicles, sweat glands, sweat ducts, and arterioles—where C-fiber endings normally terminate. Similar micro-lesions were detected in hind paw skin of mice with conditionally-induced SC Nf1-/- mutations. Hypothesizing that these microlesions were pre-cNF origins of cNF, we subsequently analyzed numerous overt, small cNF (s-cNF, 3–6 mm) and discovered that each had an adnexal structure at the epicenter of vastly increased nonpeptidergic C-fiber terminals, accompanied by excessive nSC. The IF and functional genomics assays indicated that neurturin (NTRN) and artemin (ARTN) signaling through cRET kinase and GFRα2 and GFRα3 co-receptors on the aberrant C-fiber endings and nSC may mutually promote the onset of pre-cNF and their evolution to s-cNF. Moreover, TrpA1 and TrpV1 receptors may, respectively, mediate symptoms of chronic itch and pain. These newly discovered molecular characteristics might be targeted to suppress the development of cNF and to treat chronic itch and pain symptoms in NF1 patients.
Collapse
|
23
|
GDNF pretreatment overcomes Schwann cell phenotype mismatch to promote motor axon regeneration via sensory graft. Exp Neurol 2019; 318:258-266. [PMID: 31100319 DOI: 10.1016/j.expneurol.2019.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 12/11/2022]
Abstract
In the clinic, severe motor nerve injury is commonly repaired by autologous sensory nerve bridging, but the ability of Schwann cells (SCs) in sensory nerves to support motor neuron axon growth is poor due to phenotype mismatch. In vitro experiments have demonstrated that sensory-derived SCs overcome phenotypic mismatch-induced growth inhibition after pretreatment with exogenous glial cell-derived neurotrophic factor (GDNF) and induce motor neuron axonal growth. Thus, we introduced a novel staging surgery: In the first stage of surgery, the denervated sensory nerve was pretreated with sustained-release GDNF, which was encapsulated into a self-assembling peptide nanofiber scaffold (SAPNS) RADA-16I in the donor area in vivo. In the second stage of surgery, the pretreated sensory grafts were transplanted to repair motor nerve injury. Motor axon regeneration and remyelination and muscle functional recovery after the second surgery was compared to those in the control groups. The expression of genes previously shown to be differently expressed in motor and sensory SCs was also analyzed in pretreated sensory grafts by qRT-PCR to explore possible changes after exogenous GDNF application. Exogenous GDNF acted directly on the denervated sensory nerve graft in vivo, increasing the expression of endogenous GDNF and sensory SC-derived marker brain-derived neurotrophic factor (BDNF). After transplantation to repair motor nerve injury, exogenous GDNF pretreatment promoted the regeneration and remyelination of proximal motor axons and the recovery of muscle function. Further research into how phenotype, gene expression and changes in neurotrophic factors in SCs are affected by GDNF will help us design more effective methods to treat peripheral nerve injury.
Collapse
|
24
|
Wang ZZ, Wood MD, Mackinnon SE, Sakiyama-Elbert SE. A microfluidic platform to study the effects of GDNF on neuronal axon entrapment. J Neurosci Methods 2018; 308:183-191. [PMID: 30081039 DOI: 10.1016/j.jneumeth.2018.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND One potential treatment strategy to enhance axon regeneration is transplanting Schwann Cells (SCs) that overexpress glial cell line-derived neurotrophic factor (GDNF). Unfortunately, constitutive GDNF overexpression in vivo can result in failure of regenerating axons to extend beyond the GDNF source, a phenomenon termed the "candy-store" effect. Little is known about the mechanism of this axon entrapment in vivo. NEW METHOD We present a reproducible in vitro culture platform using a microfluidic device to model axon entrapment and investigate mechanisms by which GDNF causes axon entrapment. The device is comprised of three culture chambers connected by two sets of microchannels, which prevent cell soma from moving between chambers but allow neurites to grow between chambers. Neurons from dorsal root ganglia were seeded in one end chamber while the effect of different conditions in the other two chambers was used to study neurite entrapment. RESULTS The results showed that GDNF-overexpressing SCs (G-SCs) can induce axon entrapment in vitro. We also found that while physiological levels of GDNF (100 ng/mL) promoted neurite extension, supra-physiological levels of GDNF (700 ng/mL) induced axon entrapment. COMPARISON WITH EXISTING METHOD All previous work related to the "candy-store" effect were done in vivo. Here, we report the first in vitro platform that can recapitulate the axonal entrapment and investigate the mechanism of the phenomenon. CONCLUSIONS This platform facilitates investigation of the "candy-store" effect and shows the effects of high GDNF concentrations on neurite outgrowth.
Collapse
Affiliation(s)
- Ze Zhong Wang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Matthew D Wood
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Susan E Mackinnon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | | |
Collapse
|
25
|
Influence of Genetically Modified Human Umbilical Cord Blood Mononuclear Cells on the Expression of Schwann Cell Molecular Determinants in Spinal Cord Injury. Stem Cells Int 2018. [PMID: 29531538 PMCID: PMC5835253 DOI: 10.1155/2018/4695275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Spinal cord injury (SCI) unavoidably results in death of not only neurons but also glial cells. In particular, the death of oligodendrocytes leads to impaired nerve impulse conduction in intact axons. However, after SCI, the Schwann cells (SCs) are capable of migrating towards an area of injury and participating in the formation of functional myelin. In addition to SCI, cell-based therapy can influence the migration of SCs and the expression of their molecular determinants. In a number of cases, it can be explained by the ability of implanted cells to secrete neurotrophic factors (NTFs). Genetically modified stem and progenitor cells overexpressing NTFs have recently attracted special attention of researchers and are most promising for the purposes of regenerative medicine. Therefore, we have studied the effect of genetically modified human umbilical cord blood mononuclear cells on the expression of SC molecular determinants in SCI.
Collapse
|
26
|
Postinjury Induction of Activated ErbB2 Selectively Hyperactivates Denervated Schwann Cells and Promotes Robust Dorsal Root Axon Regeneration. J Neurosci 2017; 37:10955-10970. [PMID: 28982707 DOI: 10.1523/jneurosci.0903-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 09/20/2017] [Accepted: 09/27/2017] [Indexed: 11/21/2022] Open
Abstract
Following nerve injury, denervated Schwann cells (SCs) convert to repair SCs, which enable regeneration of peripheral axons. However, the repair capacity of SCs and the regenerative capacity of peripheral axons are limited. In the present studies we examined a potential therapeutic strategy to enhance the repair capacity of SCs, and tested its efficacy in enhancing regeneration of dorsal root (DR) axons, whose regenerative capacity is particularly weak. We used male and female mice of a doxycycline-inducible transgenic line to induce expression of constitutively active ErbB2 (caErbB2) selectively in SCs after DR crush or transection. Two weeks after injury, injured DRs of induced animals contained far more SCs and SC processes. These SCs had not redifferentiated and continued to proliferate. Injured DRs of induced animals also contained far more axons that regrew along SC processes past the transection or crush site. Remarkably, SCs and axons in uninjured DRs remained quiescent, indicating that caErbB2 enhanced regeneration of injured DRs, without aberrantly activating SCs and axons in intact nerves. We also found that intraspinally expressed glial cell line-derived neurotrophic factor (GDNF), but not the removal of chondroitin sulfate proteoglycans, greatly enhanced the intraspinal migration of caErbB2-expressing SCs, enabling robust penetration of DR axons into the spinal cord. These findings indicate that SC-selective, post-injury activation of ErbB2 provides a novel strategy to powerfully enhance the repair capacity of SCs and axon regeneration, without substantial off-target damage. They also highlight that promoting directed migration of caErbB2-expressing SCs by GDNF might be useful to enable axon regrowth in a non-permissive environment.SIGNIFICANCE STATEMENT Repair of injured peripheral nerves remains a critical clinical problem. We currently lack a therapy that potently enhances axon regeneration in patients with traumatic nerve injury. It is extremely challenging to substantially increase the regenerative capacity of damaged nerves without deleterious off-target effects. It was therefore of great interest to discover that caErbB2 markedly enhances regeneration of damaged dorsal roots, while evoking little change in intact roots. To our knowledge, these findings are the first demonstration that repair capacity of denervated SCs can be efficaciously enhanced without altering innervated SCs. Our study also demonstrates that oncogenic ErbB2 signaling can be activated in SCs but not impede transdifferentiation of denervated SCs to regeneration-promoting repair SCs.
Collapse
|
27
|
The Actin Cytoskeleton Is Involved in Glial Cell Line-Derived Neurotrophic Factor (GDNF)-Induced Ret Translocation into Lipid Rafts in Dopaminergic Neuronal Cells. Int J Mol Sci 2017; 18:ijms18091922. [PMID: 28880247 PMCID: PMC5618571 DOI: 10.3390/ijms18091922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 01/04/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF), a potential therapeutic factor for Parkinson’s disease (PD), exerts its biological effects through the Ret receptor tyrosine kinase. The redistribution of Ret into lipid rafts substantially influences Ret signaling, but the mechanisms underlying Ret translocation remain unclear. The purpose of our study was to further explore the signaling mechanisms of GDNF and to determine whether the actin cytoskeleton is involved in the GDNF-induced Ret translocation into lipid rafts. In MN9D dopaminergic neuronal cells, we used density gradient centrifugation and immunofluorescence confocal microscopy to separate and visualize lipid rafts, co-immunoprecipitation to analyze protein-protein interactions, and latrunculin B (Lat B) and jasplakinolide (Jas) to disrupt and enhance the polymerization of the actin cytoskeleton, respectively. The results showed that Ret translocated into lipid rafts and coimmunoprecipitated with actin in response to GDNF treatment. After Lat B or Jas treatment, the Ret–F-actin association induced by GDNF was impaired or enhanced respectively and then the levels of Ret translocated into lipid rafts were correspondingly inhibited or promoted. These data indicate that actin polymerization and cytoskeletal remodeling are integral to GDNF-induced cell signaling in dopaminergic cells and define a new role of the actin cytoskeleton in promoting Ret redistribution into lipid rafts.
Collapse
|
28
|
Ee X, Yan Y, Hunter DA, Schellhardt L, Sakiyama-Elbert SE, Mackinnon SE, Wood MD. Transgenic SCs expressing GDNF-IRES-DsRed impair nerve regeneration within acellular nerve allografts. Biotechnol Bioeng 2017; 114:2121-2130. [PMID: 28481001 DOI: 10.1002/bit.26335] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/01/2017] [Accepted: 05/05/2017] [Indexed: 11/11/2022]
Abstract
Providing temporally regulated glial cell line-derived neurotrophic factor (GDNF) to injured nerve can promote robust axon regeneration. However, it is poorly understood why providing highly elevated levels of GDNF to nerve can lead to axon entrapment in the zone containing elevated GDNF. This limited understanding represents an obstacle to the translation of GDNF therapies to treat nerve injuries clinically. Here, we investigated how transgenic Schwann cells (SCs) overexpressing GDNF-IRES-DsRed impact nerve regeneration. Cultured primary SCs were transduced with lentiviruses (GDNF-overexpressing transgenic SCs), one of which provides the capability to express high levels of GDNF and regulate temporal GDNF expression. These SC groups were transplanted into acellular nerve allografts (ANAs) bridging a 14 mm rat sciatic nerve defect. GDNF-overexpressing transgenic SCs expressing GDNF for as little as 1 week decreased axon regeneration across ANAs and caused extensive extracellular matrix (ECM) remodeling. To determine whether additional gene expression changes beyond GDNF transgene expression occurred in GDNF-overexpressing transgenic SCs, microarray analysis of GDNF-overexpressing transgenic SCs compared to untreated SCs was performed. Microarray analysis revealed a set of common genes regulated in transgenic SC groups expressing high levels of GDNF compared to untreated SCs. A co-culture model of GDNF-overexpressing transgenic SCs with fibroblasts (FBs) revealed differential FB ECM-related gene expression compared to untreated SCs. These data suggest a component of axon entrapment is independent of GDNF's impact on axons. Biotechnol. Bioeng. 2017;114: 2121-2130. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xueping Ee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110
| | - Ying Yan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110
| | - Daniel A Hunter
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110
| | - Lauren Schellhardt
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110
| | - Shelly E Sakiyama-Elbert
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110.,Department of Biomedical Engineering, Washington University, St. Louis, Missouri.,Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Susan E Mackinnon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110
| | - Matthew D Wood
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, Missouri, 63110
| |
Collapse
|
29
|
Lu Y, Gao H, Zhang M, Chen B, Yang H. Glial Cell Line-Derived Neurotrophic Factor-Transfected Placenta-Derived Versus Bone Marrow-Derived Mesenchymal Cells for Treating Spinal Cord Injury. Med Sci Monit 2017; 23:1800-1811. [PMID: 28408732 PMCID: PMC5400030 DOI: 10.12659/msm.902754] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background Placenta-derived mesenchymal stem cells (PMSCs) were isolated from placenta and had differentiation and self-renewal potential. We transfected PMSCs with glial cell line-derived neurotrophic factor (GDNF) and compared their effect for repairing spinal cord injury (SCI) with that of GDNF-transfected bone marrow-derived mesenchymal stem cell (BMSC). Material/Methods The PMSCs were isolated from Sprague-Dawley rat placenta; BMSCs were isolated from Sprague-Dawley rat thigh bone marrow. Primary cultured BMSCs and PMSCs were uniformly spindle-shaped. Flow cytometry indicated that both cell types were CD29- and CD90-positive and CD34- and CD45-negative, confirming that they were MSCs. The PMSCs and BMSCs were transfected with recombinant lentivirus containing the GDNF gene in vitro. PMSC and BMSC viability was increased after transfection, and GDNF expression was increased until 10 d after transfection. SCI was created in the rats (n=64) and was repaired using transfected PMSCs and BMSCs or untransfected PMSCs and BMSCs. Results The transfected PMSCs and BMSCs repaired the SCI. Flow cytometry, histology, immunohistochemical, kinesiology properties, and Basso-Beattie-Bresnahan locomotion score measurements determined no significant difference between transfected PMSCs and BMSCs at 7, 14, and 21 d post-transplantation (P>0.05); the injury healed better in transfected PMSCs and BMSCs than in untransfected PMSCs and BMSCs (P<0.05). Conclusions MSCs have similar biology characteristics and capacity for SCI repair to BMSCs and can be used as a new resource for treating SCI.
Collapse
Affiliation(s)
- Yao Lu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| | - Hui Gao
- Department of Dermatology, College of Clinical of Yangzhou University, Yangzhou, Jiangsu, China (mainland)
| | - Man Zhang
- Department of Dermatology, College of Clinical of Yangzhou University, Yangzhou, Jiangsu, China (mainland)
| | - Bing Chen
- Comparative Medicine Center, Yangzhou University, Yangzhou, Jiangsu, China (mainland)
| | - Huilin Yang
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| |
Collapse
|
30
|
Electrophysiological, Morphological, and Ultrastructural Features of the Injured Spinal Cord Tissue after Transplantation of Human Umbilical Cord Blood Mononuclear Cells Genetically Modified with the VEGF and GDNF Genes. Neural Plast 2017; 2017:9857918. [PMID: 28421147 PMCID: PMC5379091 DOI: 10.1155/2017/9857918] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/24/2017] [Indexed: 01/12/2023] Open
Abstract
In this study, we examined the efficacy of human umbilical cord blood mononuclear cells (hUCB-MCs), genetically modified with the VEGF and GDNF genes using adenoviral vectors, on posttraumatic regeneration after transplantation into the site of spinal cord injury (SCI) in rats. Thirty days after SCI, followed by transplantation of nontransduced hUCB-MCs, we observed an improvement in H (latency period, LP) and M(Amax) waves, compared to the group without therapy after SCI. For genetically modified hUCB-MCs, there was improvement in Amax of M wave and LP of both the M and H waves. The ratio between Amax of the H and M waves (Hmax/Mmax) demonstrated that transplantation into the area of SCI of genetically modified hUCB-MCs was more effective than nontransduced hUCB-MCs. Spared tissue and myelinated fibers were increased at day 30 after SCI and transplantation of hUCB-MCs in the lateral and ventral funiculi 2.5 mm from the lesion epicenter. Transplantation of hUCB-MCs genetically modified with the VEGF and GNDF genes significantly increased the number of spared myelinated fibers (22-fold, P > 0.01) in the main corticospinal tract compared to the nontransduced ones. HNA+ cells with the morphology of phagocytes and microglia-like cells were found as compact clusters or cell bridges within the traumatic cavities that were lined by GFAP+ host astrocytes. Our results show that hUCB-MCs transplanted into the site of SCI improved regeneration and that hUCB-MCs genetically modified with the VEGF and GNDF genes were more effective than nontransduced hUCB-MCs.
Collapse
|
31
|
Yu Z, Men Y, Dong P. Schwann cells promote the capability of neural stem cells to differentiate into neurons and secret neurotrophic factors. Exp Ther Med 2017; 13:2029-2035. [PMID: 28565804 PMCID: PMC5443174 DOI: 10.3892/etm.2017.4183] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/20/2017] [Indexed: 11/05/2022] Open
Abstract
The present study investigated whether co-culturing Schwann cells (SCs) with neural stem cells (NSCs) improves viability, direction of differentiation and secretion of brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) in NSCs. The three groups assessed were as follows: SCs, NSCs, and a co-culture of SCs and NSCs. Cellular morphological changes were observed under an inverted phase contrast microscope and quantified. Cells were identified by immunofluorescence staining: S100 for SCs, Nestin for NSCs, microtubule associated protein (Map) 2 and NeuN for neurons and glial fibrillary acidic protein for astrocytes. Cell viability was evaluated by MTT assay. Secretion of BDNF and GDNF was quantified; mRNA expression was quantified by reverse transcription-quantitative polymerase chain reaction. The majority of NSCs in the co-cultured group differentiated into neurons. The cell survival rate of the co-culture group was significantly higher than the other groups on days 3, 5 and 10 (P<0.01). The secretion of BDNF in the co-culture group was significantly higher than NSCs on days 3, 5 and 7 (P<0.05), while the amount of GDNF in co-culture was significantly higher than both NSCs and SCs on day 1 (P<0.05). BDNF and GDNF gene expression in the co-culture group was significantly higher than SCs (P<0.01). Gene expression of Map2 in co-culture group was also significantly higher than both NSC and SC groups (P<0.01). Therefore, co-cultured SCs and NSCs promote differentiation of NSCs into neurons and secrete higher levels of neurotropic factors including BDNF and GDNF.
Collapse
Affiliation(s)
- Ziwei Yu
- Department of Otolaryngology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Yongzhi Men
- Department of Otolaryngology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Pin Dong
- Department of Otolaryngology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
32
|
Yoshizawa H, Senda D, Natori Y, Tanaka R, Mizuno H, Hayashi A. End-to-Side Neurorrhaphy as Schwann Cells Provider to Acellular Nerve Allograft and Its Suitable Application. PLoS One 2016; 11:e0167507. [PMID: 27907118 PMCID: PMC5132318 DOI: 10.1371/journal.pone.0167507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/15/2016] [Indexed: 11/23/2022] Open
Abstract
Axonal regeneration relies on support from proliferating host Schwann cells (SCs), and previous studies on acellular nerve allografts (ANGs) suggest that axons can regenerate into ANGs within a limited distance. Numerous studies have demonstrated that the supplementation of ANGs with exogenous factors, such as cultured SCs, stem cells, and growth factors, promote nerve regeneration in ANGs. However, there are several problems associated with their utilization. In this study, we investigated whether end-to-side (ETS) neurorrhaphy, which is an axonal provider, could be useful as an SC provider to support axonal elongation in ANGs. We found that ETS neurorrhaphy effectively promoted SC migration into ANGs when an epineurium window combined with partial neurectomy was performed, and the effectiveness increased when it was applied bilaterally. When we transplanted ANGs containing migrated SCs via ETS neurorrhaphy (hybrid ANGs) to the nerve gap, hybrid ANGs increased the number of regenerated axons and facilitated rapid axonal elongation, particularly when ETS neurorrhaphy was applied to both edges of the graft. This approach may represent a novel application of ETS neurorrhaphy and lead to the development of hybrid ANGs, making ANGs more practical in a clinical setting.
Collapse
Affiliation(s)
- Hidekazu Yoshizawa
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Daiki Senda
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuhei Natori
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Rica Tanaka
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Ayato Hayashi
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
33
|
Gao M, Lu P, Lynam D, Bednark B, Campana WM, Sakamoto J, Tuszynski M. BDNF gene delivery within and beyond templated agarose multi-channel guidance scaffolds enhances peripheral nerve regeneration. J Neural Eng 2016; 13:066011. [DOI: 10.1088/1741-2560/13/6/066011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
34
|
Effect of GDNF on Morphology, Proliferation, and Phagocytic Activity of Rat Neonatal Cortex Isolated Microglia. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0247-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Wang L, Sanford MT, Xin Z, Lin G, Lue TF. Role of Schwann cells in the regeneration of penile and peripheral nerves. Asian J Androl 2016; 17:776-82. [PMID: 25999359 PMCID: PMC4577590 DOI: 10.4103/1008-682x.154306] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Schwann cells (SCs) are the principal glia of the peripheral nervous system. The end point of SC development is the formation of myelinating and nonmyelinating cells which ensheath large and small diameter axons, respectively. They play an important role in axon regeneration after injury, including cavernous nerve injury that leads to erectile dysfunction (ED). Despite improvement in radical prostatectomy surgical techniques, many patients still suffer from ED postoperatively as surgical trauma causes traction injuries and local inflammatory changes in the neuronal microenvironment of the autonomic fibers innervating the penis resulting in pathophysiological alterations in the end organ. The aim of this review is to summarize contemporary evidence regarding: (1) the origin and development of SCs in the peripheral and penile nerve system; (2) Wallerian degeneration and SC plastic change following peripheral and penile nerve injury; (3) how SCs promote peripheral and penile nerve regeneration by secreting neurotrophic factors; (4) and strategies targeting SCs to accelerate peripheral nerve regeneration. We searched PubMed for articles related to these topics in both animal models and human research and found numerous studies suggesting that SCs could be a novel target for treatment of nerve injury-induced ED.
Collapse
Affiliation(s)
| | | | | | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA,
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA,
| |
Collapse
|
36
|
Mukhamedshina YO, Shaymardanova GF, Garanina ЕЕ, Salafutdinov II, Rizvanov АА, Islamov RR, Chelyshev YA. Adenoviral vector carrying glial cell-derived neurotrophic factor for direct gene therapy in comparison with human umbilical cord blood cell-mediated therapy of spinal cord injury in rat. Spinal Cord 2015; 54:347-59. [PMID: 26415641 DOI: 10.1038/sc.2015.161] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/10/2015] [Accepted: 08/12/2015] [Indexed: 12/31/2022]
Abstract
STUDY DESIGN Experimental study. OBJECTIVE To evaluate the treatment of spinal cord injury with glial cell-derived neurotrophic factor (GDNF) delivered using an adenoviral vector (AdV-GDNF group) in comparison with treatment performed using human umbilical cord blood mononuclear cells (UCB-MCs)-transduced with an adenoviral vector carrying the GDNF gene (UCB-MCs+AdV-GDNF group) in rat. SETTING Kazan, Russian Federation. METHODS We examined the efficacy of AdV-GDNF and UCB-MCs+AdV-GDNF therapy by conducting behavioral tests on the animals and morphometric studies on the spinal cord, performing immunofluorescence analyses on glial cells, investigating the survival and migration potential of UCB-MCs, and evaluating the expression of the recombinant GDNF gene. RESULTS At the 30th postoperative day, equal positive locomotor recovery was observed after both direct and cell-based GDNF therapy. However, after UCB-MCs-mediated GDNF therapy, the area of preserved tissue and the number of spared myelinated fibers were higher than those measured after direct GDNF gene therapy. Moreover, we observed distinct changes in the populations of glial cells; expression patterns of the specific markers for astrocytes (GFAP, S100B and AQP4), oligodendrocytes (PDGFαR and Cx47) and Schwann cells (P0) differed in various areas of the spinal cord of rats treated with AdV-GDNF and UCB-MCs+AdV-GDNF. CONCLUSION The differences detected in the AdV-GDNF and UCB-MCs+AdV-GDNF groups could be partially explained by the action of UCB-MCs. We discuss the insufficiency and the advantages of these two methods of GDNF gene delivery into the spinal cord after traumatic injury.
Collapse
Affiliation(s)
- Y O Mukhamedshina
- OpenLab Gene and Cell Technologies, Kazan (Volga Region) Federal University, Kazan, Russia
| | - G F Shaymardanova
- Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences, Kazan, Russia
| | - Е Е Garanina
- OpenLab Gene and Cell Technologies, Kazan (Volga Region) Federal University, Kazan, Russia
| | - I I Salafutdinov
- OpenLab Gene and Cell Technologies, Kazan (Volga Region) Federal University, Kazan, Russia
| | - А А Rizvanov
- OpenLab Gene and Cell Technologies, Kazan (Volga Region) Federal University, Kazan, Russia
| | - R R Islamov
- Department of Hystology, Kazan State Medical University, Kazan, Russia
| | - Y A Chelyshev
- Department of Hystology, Kazan State Medical University, Kazan, Russia
| |
Collapse
|
37
|
Marquardt LM, Sakiyama-Elbert SE. GDNF preconditioning can overcome Schwann cell phenotypic memory. Exp Neurol 2014; 265:1-7. [PMID: 25496841 DOI: 10.1016/j.expneurol.2014.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 11/25/2014] [Accepted: 12/03/2014] [Indexed: 01/26/2023]
Abstract
While it is known that Schwann cells (SCs) provide cues to enhance regeneration following peripheral nerve injury, the effect of SC phenotypic memory (muscle or cutaneous nerve-derived) on enhancing axonal regeneration and functional recovery has been unclear in the literature. In particular, differences between muscle and cutaneous nerve-derived SC may encourage specific motor or sensory axonal guidance in cell/tissue transplantation therapies. Thus, the goal of this study was to determine whether phenotypically matched combinations of neurons and SCs stimulate greater axonal extension compared to mismatched combinations (i.e. motor neurons/muscle nerve-derived SCs vs. motor neurons/cutaneous nerve-derived SCs). Additionally, the effect of glial cell line-derived neurotrophic factor (GDNF) treatment on SC-neuron interaction was also evaluated. In order to examine these interactions, microfluidic devices were used to assess the effects of soluble factors secreted from SCs on neurons. Unlike traditional co-culture methods, the devices allow for easier quantification of single neurite extension over long periods of time, as well as easy cell and media sampling of pure populations for biochemical analyses. Results demonstrated longer neurite growth when neurons are cultured with phenotype matched SCs, suggesting that SCs are capable of retaining phenotypic memory despite a prolonged absence of axonal contact. Furthermore, the negative effect of mismatched cultures can be overcome when mismatched SCs are preconditioned with GDNF. These results suggest that treatment of SCs with GDNF could enhance their ability to promote regeneration through mismatched grafts frequently used in clinical settings.
Collapse
Affiliation(s)
- Laura M Marquardt
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA; Division of Plastic and Reconstructive Surgery, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
38
|
Sivak WN, White JD, Bliley JM, Tien LW, Liao HT, Kaplan DL, Marra KG. Delivery of chondroitinase ABC and glial cell line-derived neurotrophic factor from silk fibroin conduits enhances peripheral nerve regeneration. J Tissue Eng Regen Med 2014; 11:733-742. [PMID: 25424415 DOI: 10.1002/term.1970] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/18/2014] [Accepted: 10/27/2014] [Indexed: 01/26/2023]
Abstract
Nerve conduits are a proven strategy for guiding axon regrowth following injury. This study compares degradable silk-trehalose films containing chondroitinase ABC (ChABC) and/or glial cell line-derived neurotrophic factor (GDNF) loaded within a silk fibroin-based nerve conduit in a rat sciatic nerve defect model. Four groups of silk conduits were prepared, with the following silk-trehalose films inserted into the conduit: (a) empty; (b) 1 µg GDNF; (3) 2 U ChABC; and (4) 1 µg GDNF/2 U ChABC. Drug release studies demonstrated 20% recovery of GDNF and ChABC at 6 weeks and 24 h, respectively. Six conduits of each type were implanted into 15 mm sciatic nerve defects in Lewis rats; conduits were explanted for histological analysis at 6 weeks. Tissues stained with Schwann cell S-100 antibody demonstrated an increased density of cells in both GDNF- and ChABC-treated groups compared to empty control conduits (p < 0.05). Conduits loaded with GDNF and ChABC also demonstrated higher levels of neuron-specific PGP 9.5 protein when compared to controls (p < 0.05). In this study we demonstrated a method to enhance Schwann cell migration and proliferation and also foster axonal regeneration when repairing peripheral nerve gap defects. Silk fibroin-based nerve conduits possess favourable mechanical and degradative properties and are further enhanced when loaded with ChABC and GDNF. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Wesley N Sivak
- Department of Plastic Surgery, University of Pittsburgh, PA, USA
| | - James D White
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | | | - Lee W Tien
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | - Han Tsung Liao
- Department of Plastic Surgery, University of Pittsburgh, PA, USA
- Department of Plastic and Reconstructive Surgery, Craniofacial Research Centre, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taiwan
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Boston, MA, USA
| | - Kacey G Marra
- Department of Plastic Surgery, University of Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
39
|
Abstract
We have recently developed aged cortical neuron cultures from autopsied human brains with Alzheimer's disease (AD). During the culturing process, we found that glutamatergic cortical neurons from the AD brain lacked a response to glial cell line-derived neurotrophic factor (GDNF), including no axonal regrowth, and were starting to undergo apoptosis. Here we showed that, in cortical neurons from age- and gender-matched cognitively normal control (NC) subjects (NC neurons), GDNF enhanced the expression of GDNF family receptor subtype α1 (GFRα1), but not the other three subtypes (GFRα2, GFRα3, and GFRα4), whereas GDNF failed to induce GFRα1 expression in cortical neurons from the AD brain (AD neurons). The exogenous introduction of GFRα1, but not of its binding partner α1-neural cell adhesion molecule, or RET into AD neurons restored the effect of GDNF on neuronal survival. Moreover, between NC and AD neurons, the AMPA receptor blocker CNQX and the NMDA receptor blocker AP-5 had opposite effects on the GFRα1 expression induced by GDNF. In NC neurons, the presence of glutamate receptors was necessary for GDNF-linked GFRα1 expression, while in AD neurons the absence of glutamate receptors was required for GFRα1 expression by GDNF stimulation. These results suggest that, in AD neurons, specific impairments of GFRα1, which may be linked to glutamatergic neurotransmission, shed light on developing potential therapeutic strategies for AD by upregulation of GFRα1 expression.
Collapse
|
40
|
Hoyng SA, De Winter F, Gnavi S, de Boer R, Boon LI, Korvers LM, Tannemaat MR, Malessy MJ, Verhaagen J. A comparative morphological, electrophysiological and functional analysis of axon regeneration through peripheral nerve autografts genetically modified to overexpress BDNF, CNTF, GDNF, NGF, NT3 or VEGF. Exp Neurol 2014; 261:578-93. [DOI: 10.1016/j.expneurol.2014.08.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 01/21/2023]
|
41
|
Moloney EB, de Winter F, Verhaagen J. ALS as a distal axonopathy: molecular mechanisms affecting neuromuscular junction stability in the presymptomatic stages of the disease. Front Neurosci 2014; 8:252. [PMID: 25177267 PMCID: PMC4132373 DOI: 10.3389/fnins.2014.00252] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/29/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is being redefined as a distal axonopathy, in that many molecular changes influencing motor neuron degeneration occur at the neuromuscular junction (NMJ) at very early stages of the disease prior to symptom onset. A huge variety of genetic and environmental causes have been associated with ALS, and interestingly, although the cause of the disease can differ, both sporadic and familial forms of ALS show a remarkable similarity in terms of disease progression and clinical manifestation. The NMJ is a highly specialized synapse, allowing for controlled signaling between muscle and nerve necessary for skeletal muscle function. In this review we will evaluate the clinical, animal experimental and cellular/molecular evidence that supports the idea of ALS as a distal axonopathy. We will discuss the early molecular mechanisms that occur at the NMJ, which alter the functional abilities of the NMJ. Specifically, we focus on the role of axon guidance molecules on the stability of the cytoskeleton and how these molecules may directly influence the cells of the NMJ in a way that may initiate or facilitate the dismantling of the neuromuscular synapse in the presymptomatic stages of ALS.
Collapse
Affiliation(s)
- Elizabeth B. Moloney
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and ScienceAmsterdam, Netherlands
| | - Fred de Winter
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and ScienceAmsterdam, Netherlands
- Department of Neurosurgery, Leiden University Medical CentreLeiden, Netherlands
| | - Joost Verhaagen
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and ScienceAmsterdam, Netherlands
- Centre for Neurogenomics and Cognitive Research, Vrije Universiteit AmsterdamAmsterdam, Netherlands
| |
Collapse
|
42
|
Ramburrun P, Kumar P, Choonara YE, Bijukumar D, du Toit LC, Pillay V. A review of bioactive release from nerve conduits as a neurotherapeutic strategy for neuronal growth in peripheral nerve injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:132350. [PMID: 25143934 PMCID: PMC4131113 DOI: 10.1155/2014/132350] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 05/04/2014] [Indexed: 02/07/2023]
Abstract
Peripheral nerve regeneration strategies employ the use of polymeric engineered nerve conduits encompassed with components of a delivery system. This allows for the controlled and sustained release of neurotrophic growth factors for the enhancement of the innate regenerative capacity of the injured nerves. This review article focuses on the delivery of neurotrophic factors (NTFs) and the importance of the parameters that control release kinetics in the delivery of optimal quantities of NTFs for improved therapeutic effect and prevention of dose dumping. Studies utilizing various controlled-release strategies, in attempt to obtain ideal release kinetics, have been reviewed in this paper. Release strategies discussed include affinity-based models, crosslinking techniques, and layer-by-layer technologies. Currently available synthetic hollow nerve conduits, an alternative to the nerve autografts, have proven to be successful in the bridging and regeneration of primarily the short transected nerve gaps in several patient cases. However, current research emphasizes on the development of more advanced nerve conduits able to simulate the effectiveness of the autograft which includes, in particular, the ability to deliver growth factors.
Collapse
Affiliation(s)
- Poornima Ramburrun
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Divya Bijukumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Lisa C. du Toit
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
43
|
Jesuraj NJ, Marquardt LM, Kwasa JA, Sakiyama-Elbert SE. Glial cell line-derived neurotrophic factor promotes increased phenotypic marker expression in femoral sensory and motor-derived Schwann cell cultures. Exp Neurol 2014; 257:10-8. [PMID: 24731946 PMCID: PMC4065822 DOI: 10.1016/j.expneurol.2014.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/06/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
Schwann cells (SCs) secrete growth factors and extracellular matrix molecules that promote neuronal survival and help guide axons during regeneration. Transplantation of SCs is a promising strategy for enhancing peripheral nerve regeneration. However, we and others have shown that after long-term in vitro expansion, SCs revert to a de-differentiated state similar to the phenotype observed after injury. In vivo, glial cell-line derived neurotrophic factor (GDNF) may guide the differentiation of SCs to remyelinate regenerating axons. Therefore, we hypothesized that exogenous GDNF may guide the differentiation of SCs into their native phenotypes in vitro through stimulation of GDNF family receptor (GFR)α-1. When activated in SCs, GFRα-1 promotes phosphorylation of Fyn, a Src family tyrosine kinase responsible for mediating downstream signaling for differentiation and proliferation. In this study, SCs harvested from the sensory and motor branches of rat femoral nerve were expanded in vitro and then cultured with 50 or 100ng/mL of GDNF. The exogenous GDNF promoted differentiation of sensory and motor-derived SCs back to their native phenotypes, as demonstrated by decreased proliferation after 7days and increased expression of S100Ββ and phenotype-specific markers. Furthermore, inhibiting Fyn with Src family kinase inhibitors, PP2 and SU6656, and siRNA-mediated knockdown of Fyn reduced GDNF-stimulated differentiation of sensory and motor-derived SCs. These results demonstrate that activating Fyn is necessary for GDNF-stimulated differentiation of femoral nerve-derived SCs into their native phenotypes in vitro. Therefore GDNF could be incorporated into SC-based therapies to promote differentiation of SCs into their native phenotype to improve functional nerve regeneration.
Collapse
Affiliation(s)
- Nithya J Jesuraj
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Laura M Marquardt
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Jasmine A Kwasa
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
44
|
Hegarty SV, Collins LM, Gavin AM, Roche SL, Wyatt SL, Sullivan AM, O'Keeffe GW. Canonical BMP-Smad signalling promotes neurite growth in rat midbrain dopaminergic neurons. Neuromolecular Med 2014; 16:473-89. [PMID: 24682653 DOI: 10.1007/s12017-014-8299-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/07/2014] [Indexed: 01/01/2023]
Abstract
Ventral midbrain (VM) dopaminergic (DA) neurons project to the dorsal striatum via the nigrostriatal pathway to regulate voluntary movements, and their loss leads to the motor dysfunction seen in Parkinson's disease (PD). Despite recent progress in the understanding of VM DA neurogenesis, the factors regulating nigrostriatal pathway development remain largely unknown. The bone morphogenetic protein (BMP) family regulates neurite growth in the developing nervous system and may contribute to nigrostriatal pathway development. Two related members of this family, BMP2 and growth differentiation factor (GDF)5, have neurotrophic effects, including promotion of neurite growth, on cultured VM DA neurons. However, the molecular mechanisms regulating their effects on DA neurons are unknown. By characterising the temporal expression profiles of endogenous BMP receptors (BMPRs) in the developing and adult rat VM and striatum, this study identified BMP2 and GDF5 as potential regulators of nigrostriatal pathway development. Furthermore, through the use of noggin, dorsomorphin and BMPR/Smad plasmids, this study demonstrated that GDF5- and BMP2-induced neurite outgrowth from cultured VM DA neurons is dependent on BMP type I receptor activation of the Smad 1/5/8 signalling pathway.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
45
|
Roles for the TGFβ superfamily in the development and survival of midbrain dopaminergic neurons. Mol Neurobiol 2014; 50:559-73. [PMID: 24504901 DOI: 10.1007/s12035-014-8639-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/02/2014] [Indexed: 12/29/2022]
Abstract
The adult midbrain contains 75% of all dopaminergic neurons in the CNS. Within the midbrain, these neurons are divided into three anatomically and functionally distinct clusters termed A8, A9 and A10. The A9 group plays a functionally non-redundant role in the control of voluntary movement, which is highlighted by the motor syndrome that results from their progressive degeneration in the neurodegenerative disorder, Parkinson's disease. Despite 50 years of investigation, treatment for Parkinson's disease remains symptomatic, but an intensive research effort has proposed delivering neurotrophic factors to the brain to protect the remaining dopaminergic neurons, or using these neurotrophic factors to differentiate dopaminergic neurons from stem cell sources for cell transplantation. Most neurotrophic factors studied in this context have been members of the transforming growth factor β (TGFβ) superfamily. In recent years, an intensive research effort has focused on understanding the function of these proteins in midbrain dopaminergic neuron development and their role in the molecular architecture that regulates the development of this brain region, with the goal of applying this knowledge to develop novel therapies for Parkinson's disease. In this review, the current evidence showing that TGFβ superfamily members play critical roles in the regulation of midbrain dopaminergic neuron induction, differentiation, target innervation and survival during embryonic and postnatal development is analysed, and the implications of these findings are discussed.
Collapse
|
46
|
Jesuraj NJ, Santosa KB, Macewan MR, Moore AM, Kasukurthi R, Ray WZ, Flagg ER, Hunter DA, Borschel GH, Johnson PJ, Mackinnon SE, Sakiyama-Elbert SE. Schwann cells seeded in acellular nerve grafts improve functional recovery. Muscle Nerve 2013; 49:267-76. [PMID: 23625513 DOI: 10.1002/mus.23885] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2013] [Indexed: 12/14/2022]
Abstract
INTRODUCTION This study evaluated whether Schwann cells (SCs) from different nerve sources transplanted into cold-preserved acellular nerve grafts (CP-ANGs) would improve functional regeneration compared with nerve isografts. METHODS SCs isolated and expanded from motor and sensory branches of rat femoral and sciatic nerves were seeded into 14mm CP-ANGs. Growth factor expression, axonal regeneration, and functional recovery were evaluated in a 14-mm rat sciatic injury model and compared with isografts. RESULTS At 14 days, motor or sensory-derived SCs increased expression of growth factors in CP-ANGs versus isografts. After 42 days, histomorphometric analysis found CP-ANGs with SCs and isografts had similar numbers of regenerating nerve fibers. At 84 days, muscle force generation was similar for CP-ANGs with SCs and isografts. SC source did not affect nerve fiber counts or muscle force generation. CONCLUSIONS SCs transplanted into CP-ANGs increase functional regeneration to isograft levels; however SC nerve source did not have an effect.
Collapse
Affiliation(s)
- Nithya J Jesuraj
- Department of Biomedical Engineering, Washington University, Campus Box 1097, One Brookings Drive, St. Louis, Missouri, 63130, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lentiviral vector-mediated gradients of GDNF in the injured peripheral nerve: effects on nerve coil formation, Schwann cell maturation and myelination. PLoS One 2013; 8:e71076. [PMID: 23951085 PMCID: PMC3741360 DOI: 10.1371/journal.pone.0071076] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/01/2013] [Indexed: 12/24/2022] Open
Abstract
Although the peripheral nerve is capable of regeneration, only a small minority of patients regain normal function after surgical reconstruction of a major peripheral nerve lesion, resulting in a severe and lasting negative impact on the quality of life. Glial cell-line derived neurotrophic factor (GDNF) has potent survival- and outgrowth-promoting effects on motoneurons, but locally elevated levels of GDNF cause trapping of regenerating axons and the formation of nerve coils. This phenomenon has been called the “candy store” effect. In this study we created gradients of GDNF in the sciatic nerve after a ventral root avulsion. This approach also allowed us to study the effect of increasing concentrations of GDNF on Schwann cell proliferation and morphology in the injured peripheral nerve. We demonstrate that lentiviral vectors can be used to create a 4 cm long GDNF gradient in the intact and lesioned rat sciatic nerve. Nerve coils were formed throughout the gradient and the number and size of the nerve coils increased with increasing GDNF levels in the nerve. In the nerve coils, Schwann cell density is increased, their morphology is disrupted and myelination of axons is severely impaired. The total number of regenerated and surviving motoneurons is not enhanced after the distal application of a GDNF gradient, but increased sprouting does result in higher number of motor axon in the distal segment of the sciatic nerve. These results show that lentiviral vector mediated overexpression of GDNF exerts multiple effects on both Schwann cells and axons and that nerve coil formation already occurs at relatively low concentrations of exogenous GDNF. Controlled expression of GDNF, by using a viral vector with regulatable GDNF expression, may be required to avoid motor axon trapping and to prevent the effects on Schwann cell proliferation and myelination.
Collapse
|
48
|
Synergistic Lithium Chloride and Glial Cell Line–Derived Neurotrophic Factor Delivery for Peripheral Nerve Repair in a Rodent Sciatic Nerve Injury Model. Plast Reconstr Surg 2013; 132:251e-262e. [DOI: 10.1097/prs.0b013e31829588cf] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Ansorena E, De Berdt P, Ucakar B, Simón-Yarza T, Jacobs D, Schakman O, Jankovski A, Deumens R, Blanco-Prieto MJ, Préat V, des Rieux A. Injectable alginate hydrogel loaded with GDNF promotes functional recovery in a hemisection model of spinal cord injury. Int J Pharm 2013; 455:148-58. [PMID: 23916821 DOI: 10.1016/j.ijpharm.2013.07.045] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/15/2013] [Accepted: 07/17/2013] [Indexed: 11/29/2022]
Abstract
We hypothesized that local delivery of GDNF in spinal cord lesion via an injectable alginate hydrogel gelifying in situ would support spinal cord plasticity and functional recovery. The GDNF release from the hydrogel was slowed by GDNF encapsulation in microspheres compared to non-formulated GDNF (free GDNF). When injected in a rat spinal cord hemisection model, more neurofilaments were observed in the lesion when the rats were treated with free GDNF-loaded hydrogels. More growing neurites were detected in the tissues surrounding the lesion when the animals were treated with GDNF microsphere-loaded hydrogels. Intense GFAP (astrocytes), low βIII tubulin (neural cells) and RECA-1 (endothelial cells) stainings were observed for non-treated lesions while GDNF-treated spinal cords presented less GFAP staining and more endothelial and nerve fiber infiltration in the lesion site. The animals treated with free GDNF-loaded hydrogel presented superior functional recovery compared with the animals treated with the GDNF microsphere-loaded hydrogels and non-treated animals.
Collapse
Affiliation(s)
- Eduardo Ansorena
- Université Catholique de Louvain, Louvain Drug Research Institute, Pharmaceutics and Drug delivery Unit, 1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chen CH, Huang SY, Chen NF, Feng CW, Hung HC, Sung CS, Jean YH, Wen ZH, Chen WF. Intrathecal granulocyte colony-stimulating factor modulate glial cell line-derived neurotrophic factor and vascular endothelial growth factor A expression in glial cells after experimental spinal cord ischemia. Neuroscience 2013; 242:39-52. [DOI: 10.1016/j.neuroscience.2013.02.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/30/2013] [Accepted: 02/09/2013] [Indexed: 12/20/2022]
|