1
|
Pan Y, Lin H, Chung M, Yang Y, Zhang L, Pan X, Cai S. Generation of phenotypically stable and functionally mature human bone marrow MSCs derived Schwann cells via the induction of human iPSCs-derived sensory neurons. Stem Cell Res Ther 2025; 16:106. [PMID: 40025574 PMCID: PMC11872329 DOI: 10.1186/s13287-025-04217-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Phenotypically unstable Schwann cell-like cells (SCLCs), derived from mesenchymal stem cells (MSCs) require intercellular contact-mediated cues for Schwann cell (SCs)-fate commitment. Although rat dorsal root ganglion (DRG) neurons provide contact-mediated signals for the conversion of SCLCs into fate-committed SCs, the use of animal cells is clinically unacceptable. To overcome this problem, we previously acquired human induced pluripotent stem cell-derived sensory neurons (hiPSC-dSNs) as surrogates of rat DRG neurons that committed rat bone marrow SCLCs to the SC fate. In this study, we explored whether hiPSC-dSNs could mimic rat DRG neuron effects to obtain fate-committed SCs from hBMSC-derived SCLCs. METHODS hiPSCs were induced into hiPSC-dSNs using a specific chemical small molecule combination. hBMSCs were induced into hBMSC-derived SCLCs in a specific culture medium and then co-cultured with hiPSC-dSNs to generate SCs. The identity of hBMSC-derived SCs (hBMSC-dSCs) was examined by immunofluorescence, western bolt, electronic microscopy, and RNA-seq. Immunofluorescence was also used to detect the myelination capacity. Enzyme-linked immunosorbent assay and neurite outgrowth analysis were used to test the secretion of neurotrophic factors. RESULTS The hBMSC-dSCs exhibited bi-/tri-polar morphology of SCs and maintained the expression of the SC markers S100, p75NTR, p0, GFAP, and Sox10, even after withdrawing the glia-inducing factors or hiPSC-dSNs. Electronic microscopy and RNA-seq analysis provided evidence that hBMSC-dSCs were similar to the original human SCs in terms of their function and a variety of characteristics. Furthermore, these cells formed MBP-positive segments and secreted neurotrophic factors to facilitate the neurite outgrowth of Neuro2A. CONCLUSIONS These results demonstrated that phenotypically stable and functionally mature hBMSC-dSCs were generated efficiently via the co-culture of hiPSC-dSNs and hBMSC-derived SCLCs. Our findings may provide a promising protocol through which stable and fully developed hBMSC-dSCs can be used for transplantation to regenerate myelin sheath.
Collapse
Affiliation(s)
- Yu Pan
- Laboratory of Regenerative Medicine, Medical School, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Haohui Lin
- Laboratory of Regenerative Medicine, Medical School, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Manhon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Yang
- Laboratory of Regenerative Medicine, Medical School, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Li Zhang
- Laboratory of Regenerative Medicine, Medical School, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Xiaohua Pan
- Laboratory of Regenerative Medicine, Medical School, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Sa Cai
- Laboratory of Regenerative Medicine, Medical School, The 2nd Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China.
| |
Collapse
|
2
|
Wei S, Xiong F, Gu H, Zhang Z, Xuan H, Jin Y, Xue Y, Li B, Feng W, Yuan H. Highly aligned electroactive ultrafine fibers promote the differentiation of mesenchymal stem cells into Schwann-like cells for nerve regeneration. Int J Biol Macromol 2024; 279:135388. [PMID: 39255892 DOI: 10.1016/j.ijbiomac.2024.135388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
This study investigates the efficacy of a novel tissue-engineered scaffold for nerve repair and functional reconstruction following injury. Utilizing stable jet electrospinning, we fabricated aligned ultrafine fibers from dopamine and poly(L-lactic acid) (PLLA), further developing a biomimetic, oriented, and electroactive scaffold comprising poly(pyrrole) (PPy), polydopamine (PDA), and PLLA through dual in situ polymerizations. The scaffold demonstrated enhanced cell adhesion and reactive oxygen species (ROS) scavenging capabilities and promoted the differentiation of mesenchymal stem cells (MSCs) into Schwann-like cells, essential for nerve regeneration. In vivo assessments revealed significant peripheral nerve regeneration in 10 mm sciatic nerve defects in rats, with observations made 12 weeks post-transplantation. This included facilitated myelination and increased muscle density on the injured side, leading to improved motor function recovery. Our results suggest that the aligned PPy/PDA/PLLA fibrous scaffold offers a promising approach for promoting the differentiation of MSCs into Schwann-like cells conducive to nerve regeneration and represents a significant advancement in nerve repair technologies. This study provides a foundational basis for future research into tissue-engineered solutions for nerve damage, potentially impacting clinical strategies for nerve reconstruction.
Collapse
Affiliation(s)
- Shuo Wei
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Feng Xiong
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Haonan Gu
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Zhuojun Zhang
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Hongyun Xuan
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Yan Jin
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Ye Xue
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China
| | - Biyun Li
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China.
| | - Wei Feng
- School of Life Sciences, Shanghai University, Shanghai 200444, PR China; School of Medicine, Shanghai University, Shanghai 200444, PR China.
| | - Huihua Yuan
- School of Life Sciences, Nantong University, Nantong, Jiangsu 226019, PR China.
| |
Collapse
|
3
|
Sun W, Ye B, Chen S, Zeng L, Lu H, Wan Y, Gao Q, Chen K, Qu Y, Wu B, Lv X, Guo X. Neuro-bone tissue engineering: emerging mechanisms, potential strategies, and current challenges. Bone Res 2023; 11:65. [PMID: 38123549 PMCID: PMC10733346 DOI: 10.1038/s41413-023-00302-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/08/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
The skeleton is a highly innervated organ in which nerve fibers interact with various skeletal cells. Peripheral nerve endings release neurogenic factors and sense skeletal signals, which mediate bone metabolism and skeletal pain. In recent years, bone tissue engineering has increasingly focused on the effects of the nervous system on bone regeneration. Simultaneous regeneration of bone and nerves through the use of materials or by the enhancement of endogenous neurogenic repair signals has been proven to promote functional bone regeneration. Additionally, emerging information on the mechanisms of skeletal interoception and the central nervous system regulation of bone homeostasis provide an opportunity for advancing biomaterials. However, comprehensive reviews of this topic are lacking. Therefore, this review provides an overview of the relationship between nerves and bone regeneration, focusing on tissue engineering applications. We discuss novel regulatory mechanisms and explore innovative approaches based on nerve-bone interactions for bone regeneration. Finally, the challenges and future prospects of this field are briefly discussed.
Collapse
Affiliation(s)
- Wenzhe Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bing Ye
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Siyue Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lian Zeng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hongwei Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yizhou Wan
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qing Gao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaifang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Bin Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
4
|
Delcroix GJR, Hackett A, Schiller PC, Temple HT. Characterization of three washing/decellularization procedures for the production of bioactive human micronized neural tissue (hMINT). Cell Tissue Bank 2023; 24:693-703. [PMID: 36854877 DOI: 10.1007/s10561-023-10075-3] [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: 10/25/2022] [Accepted: 01/29/2023] [Indexed: 03/02/2023]
Abstract
BACKGROUND We developed a novel, injectable and decellularized human peripheral nerve-based scaffold, named Micronized Human Neural Tissue (hMINT), designed to be used as a supportive matrix for stem cell transplantation in the context of spinal cord injury (SCI). MATERIALS AND METHODS Human donated sciatic nerves were micronized at liquid nitrogen temperature prior to decellularization using 3 different procedures of various harshness. hMINT were characterized in terms of particle size, DNA, sulfated glycosaminoglycans (sGAG) and growth factors content. To test the biocompatibility and bioactivity of the various preparations, we used a type of mesenchymal stromal cells (MSCs), termed MIAMI cells, which were placed in contact with hMINT to monitor cell attachment by confocal microscopy and gene expression by RT-qPCR in vitro. RESULTS The content of DNA, sGAG and growth factors left in the product after processing was highly dependent on the decellularization procedure used. We demonstrated that hMINT are biocompatible and promoted the attachment and long-term survival of MIAMI cells in vitro. Finally, combination with hMINT increased MIAMI cells mRNA expression of pro-survival and anti-inflammatory factors. Importantly, the strongest bioactivity on MIAMI cells was observed with the hMINT decellularized using the mildest decellularization procedure, therefore emphasizing the importance of achieving an adequate decellularization without losing the hMINT's bioactivity. PERSPECTIVES AND CLINICAL SIGNIFICANCE The capacity of hMINT/stem cells to facilitate protection of injured neural tissue, promote axon re-growth and improve functional recovery will be tested in an animal model of SCI and other neurodegenerative disorders in the future.
Collapse
Affiliation(s)
- Gaëtan J-R Delcroix
- College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA.
| | - Amber Hackett
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Paul C Schiller
- Geriatric Research Education and Clinical Center, Miami VA Healthcare System, Miami, FL, USA
| | | |
Collapse
|
5
|
Wang Z, Zheng D, Tan YS, Yuan Q, Yuan F, Zhang S. Enabling Survival of Transplanted Neural Precursor Cells in the Ischemic Brain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302527. [PMID: 37867250 PMCID: PMC10667812 DOI: 10.1002/advs.202302527] [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: 04/20/2023] [Revised: 07/24/2023] [Indexed: 10/24/2023]
Abstract
There is no effective therapy for ischemic stroke following the acute stage. Neural transplantation offers a potential option for repairing the ischemic lesion. However, this strategy is hindered by the poor survival of the neural precursor cells (NPCs) that are transplanted into the inflammatory ischemic core. Here, a chemical cocktail consisting of fibrinogen and maraviroc is developed to promote the survival of the transplanted NPCs in the ischemic core of the mouse cerebral cortex. The grafted NPCs survive in the presence of the cocktail but not fibrinogen or maraviroc alone at day 7. The surviving NPCs divide and differentiate to mature neurons by day 30, reconstituting the infarct cortex with vascularization. Molecular analysis in vivo and in vitro shows that blocking the activation of CCR5 on the NPCs protects the NPCs from apoptosis induced by pro-inflammatory factors, revealing the underlying protective effect of the cocktail for NPCs. The findings open an avenue to enable survival of the transplanted NPCs under the inflammatory neurological conditions like stroke.
Collapse
Affiliation(s)
- Zhifu Wang
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Danyi Zheng
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Ye Sing Tan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Qiang Yuan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Fang Yuan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Su‐Chun Zhang
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
- Department of NeuroscienceDepartment of NeurologyWaisman CenterUniversity of Wisconsin‐MadisonMadisonWI53705USA
| |
Collapse
|
6
|
Solomevich SO, Oranges CM, Kalbermatten DF, Schwendeman A, Madduri S. Natural polysaccharides and their derivatives as potential medical materials and drug delivery systems for the treatment of peripheral nerve injuries. Carbohydr Polym 2023; 315:120934. [PMID: 37230605 DOI: 10.1016/j.carbpol.2023.120934] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023]
Abstract
Peripheral nerve repair following injury is one of the most serious problems in neurosurgery. Clinical outcomes are often unsatisfactory and associated with a huge socioeconomic burden. Several studies have revealed the great potential of biodegradable polysaccharides for improving nerve regeneration. We review here the promising therapeutic strategies involving different types of polysaccharides and their bio-active composites for promoting nerve regeneration. Within this context, polysaccharide materials widely used for nerve repair in different forms are highlighted, including nerve guidance conduits, hydrogels, nanofibers and films. While nerve guidance conduits and hydrogels were used as main structural scaffolds, the other forms including nanofibers and films were generally used as additional supporting materials. We also discuss the issues of ease of therapeutic implementation, drug release properties and therapeutic outcomes, together with potential future directions of research.
Collapse
Affiliation(s)
- Sergey O Solomevich
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA; Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus
| | - Carlo M Oranges
- Plastic, Reconstructive and Aesthetic Surgery Division, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Daniel F Kalbermatten
- Plastic, Reconstructive and Aesthetic Surgery Division, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Bioengineering and Neuroregeneration Laboratory, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Srinivas Madduri
- Plastic, Reconstructive and Aesthetic Surgery Division, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Bioengineering and Neuroregeneration Laboratory, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
7
|
Drewry MD, Rothermund K, Syed-Picard FN. Topographical and Chemical Inductive Cues Synergistically Enhance the Schwann Cell Differentiation of Aligned Dental Pulp Stem Cell Sheets. J Tissue Eng Regen Med 2023; 2023:7958770. [PMID: 40226400 PMCID: PMC11918939 DOI: 10.1155/2023/7958770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 04/15/2025]
Abstract
Peripheral nerves have an inherent capacity for regeneration, but these Schwann cell-mediated mechanisms are insufficient for severe injuries. With current clinical treatments, slow regeneration and aberrant reinnervation result in poor functional outcomes. Dental pulp stem cells (DPSCs) offer a promising source of therapeutic neurotrophic factors (NTFs), growth factors that stimulate axon regeneration. Previously, we established that DPSCs can generate scaffold-free sheets with a linearly aligned extracellular matrix (ECM). These sheets provide trophic cues via the DPSCs and directional cues through the aligned ECM to both accelerate and orient axon outgrowth, thus providing a biomaterial capable of addressing the current clinical challenges. DPSCs have a propensity for differentiating into Schwann cells (SC-DPSCs), further enhancing their endogenous NTF expression. Here, we evaluated the effect of inducing SC differentiation on the neuroregenerative bioactivity of our DPSC sheets. These sheets were formed on substrates with linear microgrooves to direct the cells to deposit an aligned ECM. Inducing differentiation using an SC differentiation medium (SCDM) increased NTF expression 2-fold compared to unaligned uDPSC sheets, and this effect was amplified in linearly oriented SC-DPSC sheets by up to 8-fold. Furthermore, these aligned SC-DPSC sheets remodeled the sheet ECM to more closely emulate a regenerative neural microenvironment, expressing 8-fold and 2 × 107-fold more collagen IV and laminin, respectively, than unaligned uDPSC sheets. These data demonstrate that the chemical cues of the SCDM and the mechanotransductive cues of the aligned cell sheet synergistically enhanced the differentiation of DPSCs into repair SC-like cells. To evaluate their functional effects on neuritogenesis, the DPSC sheets were directly cocultured with neuronally differentiated neuroblastoma SH-SY5Y cells. In this in vitro culture system, the aligned SC-DPSC sheets promoted oriented neurite-like outgrowth similar to aligned uninduced DPSC sheets and increased collateral branching, which may emulate stages associated with natural SC-mediated repair processes. Therefore, linearly aligned SC-DPSC sheets have the potential to both promote nerve regeneration and reduce aberrant reinnervation, thus providing a promising biomaterial for applications to improve the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Michelle D. Drewry
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kristi Rothermund
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fatima N. Syed-Picard
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Lischer M, di Summa PG, Petrou IG, Schaefer DJ, Guzman R, Kalbermatten DF, Madduri S. Mesenchymal Stem Cells in Nerve Tissue Engineering: Bridging Nerve Gap Injuries in Large Animals. Int J Mol Sci 2023; 24:ijms24097800. [PMID: 37175506 PMCID: PMC10177884 DOI: 10.3390/ijms24097800] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Cell-therapy-based nerve repair strategies hold great promise. In the field, there is an extensive amount of evidence for better regenerative outcomes when using tissue-engineered nerve grafts for bridging severe gap injuries. Although a massive number of studies have been performed using rodents, only a limited number involving nerve injury models of large animals were reported. Nerve injury models mirroring the human nerve size and injury complexity are crucial to direct the further clinical development of advanced therapeutic interventions. Thus, there is a great need for the advancement of research using large animals, which will closely reflect human nerve repair outcomes. Within this context, this review highlights various stem cell-based nerve repair strategies involving large animal models such as pigs, rabbits, dogs, and monkeys, with an emphasis on the limitations and strengths of therapeutic strategy and outcome measurements. Finally, future directions in the field of nerve repair are discussed. Thus, the present review provides valuable knowledge, as well as the current state of information and insights into nerve repair strategies using cell therapies in large animals.
Collapse
Affiliation(s)
- Mirko Lischer
- Center for Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
| | - Pietro G di Summa
- Department of Plastic, Reconstructive and Hand Surgery, University Hospital of Lausanne and University of Lausanne, 1015 Lausanne, Switzerland
| | - Ilias G Petrou
- Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, University Hospitals and University of Geneva, 1205 Geneva, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Raphael Guzman
- Department of Neurosurgery, University Hospital Basel, 4031 Basel, Switzerland
| | - Daniel F Kalbermatten
- Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, University Hospitals and University of Geneva, 1205 Geneva, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Srinivas Madduri
- Center for Bioengineering and Regenerative Medicine, Department of Biomedical Engineering, University of Basel, 4123 Allschwil, Switzerland
- Plastic, Reconstructive and Aesthetic Surgery, Department of Surgery, University Hospitals and University of Geneva, 1205 Geneva, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
- Bioengineering and Neuroregeneration, Department of Surgery, Geneva University Hospitals and University of Geneva, 1205 Geneva, Switzerland
| |
Collapse
|
9
|
Perrelle JM, Boreland AJ, Gamboa JM, Gowda P, Murthy NS. Biomimetic Strategies for Peripheral Nerve Injury Repair: An Exploration of Microarchitecture and Cellularization. BIOMEDICAL MATERIALS & DEVICES (NEW YORK, N.Y.) 2023; 1:21-37. [PMID: 38343513 PMCID: PMC10857769 DOI: 10.1007/s44174-022-00039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/14/2022] [Indexed: 02/15/2024]
Abstract
Injuries to the nervous system present formidable challenges to scientists, clinicians, and patients. While regeneration within the central nervous system is minimal, peripheral nerves can regenerate, albeit with limitations. The regenerative mechanisms of the peripheral nervous system thus provide fertile ground for clinical and scientific advancement, and opportunities to learn fundamental lessons regarding nerve behavior in the context of regeneration, particularly the relationship of axons to their support cells and the extracellular matrix environment. However, few current interventions adequately address peripheral nerve injuries. This article aims to elucidate areas in which progress might be made toward developing better interventions, particularly using synthetic nerve grafts. The article first provides a thorough review of peripheral nerve anatomy, physiology, and the regenerative mechanisms that occur in response to injury. This is followed by a discussion of currently available interventions for peripheral nerve injuries. Promising biomaterial fabrication techniques which aim to recapitulate nerve architecture, along with approaches to enhancing these biomaterial scaffolds with growth factors and cellular components, are then described. The final section elucidates specific considerations when developing nerve grafts, including utilizing induced pluripotent stem cells, Schwann cells, nerve growth factors, and multilayered structures that mimic the architectures of the natural nerve.
Collapse
Affiliation(s)
- Jeremy M. Perrelle
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Andrew J. Boreland
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Graduate Program in Molecular Biosciences, Rutgers University, Piscataway, NJ, USA
| | - Jasmine M. Gamboa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Prarthana Gowda
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - N. Sanjeeva Murthy
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
10
|
Fujiwara Y, Kusakabe KT, Baba K, Sasaki N. Effect of platelet lysate on Schwann-like cell differentiation of equine bone marrow-derived mesenchymal stem cells. Res Vet Sci 2023; 159:11-18. [PMID: 37060838 DOI: 10.1016/j.rvsc.2023.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Currently, treatment for peripheral nerve injuries in horses primarily relies upon physical therapy and anti-inflammatory drugs. In humans, various treatments using mesenchymal stem cells (MSCs) are being attempted. Therefore, in this study, Schwann-like cell differentiation cultures of equine MSCs were prepared using fetal bovine serum (FBS) and equine platelet lysate (ePL). ePL increased the platelet count to 1 × 106/μl, the optimal concentration for culture. In both groups, an elongated morphology at both ends, characteristic of Schwann cells, was observed under the microscope. Real-time PCR analysis of the expression levels of neuronal markers showed that the ePL group tended to express higher levels of Nestin, Musashi1, and Pax3 than the FBS group. p75 was expressed at low levels in both groups. Immunostaining results showed localization of Nestin in both groups of differentiated cells, but the positive cell rate was significantly higher in the ePL group than in the FBS group. Overall, the ePL gro showed good results for Schwann-like cell differentiation, which may be useful for future use in the treatment of equine motor neuron disease. This knowledge could be applied translationaly in the treatment of amyotrophic lateral sclerosis in humans.Overall, the ePL group showed good results for Schwann-like cell differentiation, which may be useful for future use in the treatment of equine motor neuron disease and in the treatment of amyotrophic lateral sclerosis in humans.
Collapse
|
11
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
12
|
Singh VK, Haq A, Tiwari M, Saxena AK. Approach to management of nerve gaps in peripheral nerve injuries. Injury 2022; 53:1308-1318. [PMID: 35105440 DOI: 10.1016/j.injury.2022.01.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 02/02/2023]
Abstract
Peripheral nerve injuries (PNI) are a major clinical problem. In general, PNI results from motor vehicle accidents, lacerations with sharp objects, penetrating trauma (gunshot wounds) and stretching or crushing trauma and fractures. They can result in significant morbidity, including motor and/or sensory loss, which can affect significantly the life of the patient. Currently, the standard surgical technique for complete nerve transection is end-to-end neurorrhaphy. Unfortunately, there is segmental loss of the nerve trunk in some cases where nerve mobilization may permit end-to-end neurorrhaphy if the gap is less than 1 cm. When the nerve gap exceeds 1 cm, autologous nerve grafting is the gold standard of treatment. But in light of limited availability and concerned donor site morbidity, other techniques have been used: vascularized nerve grafts, cellular and acellular allografts, nerve conduits, nerve transfers and end-to-side neurorrhaphy. This review intends to present an overview of the literature on the applications of these techniques in repair of peripheral nerve injuries. This article also focuses on preoperative assessment, surgical timing, available options and future perspectives.
Collapse
Affiliation(s)
- Veena K Singh
- Department of Burns & Plastic surgery, All India Institute of Medical Sciences, Patna, Bihar, India.
| | - Ansarul Haq
- Department of Burns & Plastic surgery, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Meenakshi Tiwari
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Ajit K Saxena
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences, Patna, Bihar, India
| |
Collapse
|
13
|
Govbakh I, Kyryk V, Ustymenko A, Rubtsov V, Tsupykov O, Bulgakova NV, Zavodovskiy DO, Sokolowska I, Maznychenko A. Stem Cell Therapy Enhances Motor Activity of Triceps Surae Muscle in Mice with Hereditary Peripheral Neuropathy. Int J Mol Sci 2021; 22:ijms222112026. [PMID: 34769453 PMCID: PMC8584487 DOI: 10.3390/ijms222112026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/27/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Impaired motor and sensory functions are the main features of Charcot-Marie-Tooth disease. Mesenchymal stem cell (MSCs) therapy is one of the possible treatments for this disease. It was assumed that MSCs therapy can improve the contractile properties of the triceps surae (TS) muscles in mice with hereditary peripheral neuropathy. Murine adipose-derived mesenchymal stromal cells (AD-MSCs) were obtained for transplantation into TS muscles of FVB-C-Tg(GFPU)5Nagy/J mice. Three months after AD-MSCs transplantation, animals were subjected to electrophysiological investigations. Parameters of TS muscle tension after intermittent high frequency electrical sciatic nerve stimulations were analyzed. It was found that force of TS muscle tension contraction in animals after AD-MSCs treatment was two-time higher than in untreated mice. Normalized values of force muscle contraction in different phases of electrical stimulation were 0.3 ± 0.01 vs. 0.18 ± 0.01 and 0.26 ± 0.03 vs. 0.13 ± 0.03 for treated and untreated animals, respectively. It is assumed that the two-fold increase in TS muscle strength was caused by stem cell therapy. Apparently, AD-MSCs therapy can promote nerve regeneration and partial restoration of muscle function, and thus can be a potential therapeutic agent for the treatment of peripheral neuropathies.
Collapse
Affiliation(s)
- Iryna Govbakh
- Department of General Practice-Family Medicine, Kharkiv Medical Academy of Postgraduate Education, 61000 Kharkiv, Ukraine;
| | - Vitalii Kyryk
- Cell and Tissue Technologies Department, State Institute of Genetic and Regenerative Medicine NAMS of Ukraine, 04114 Kyiv, Ukraine; (V.K.); (A.U.); (O.T.)
- Laboratory of Pathophysiology and Immunology, D. F. Chebotarev State Institute of Gerontology NAMS of Ukraine, 04114 Kyiv, Ukraine
| | - Alina Ustymenko
- Cell and Tissue Technologies Department, State Institute of Genetic and Regenerative Medicine NAMS of Ukraine, 04114 Kyiv, Ukraine; (V.K.); (A.U.); (O.T.)
- Laboratory of Pathophysiology and Immunology, D. F. Chebotarev State Institute of Gerontology NAMS of Ukraine, 04114 Kyiv, Ukraine
| | - Volodymyr Rubtsov
- Department of Cytology, Histology and Reproductive Medicine, Educational and Scientific Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, 03127 Kyiv, Ukraine;
| | - Oleg Tsupykov
- Cell and Tissue Technologies Department, State Institute of Genetic and Regenerative Medicine NAMS of Ukraine, 04114 Kyiv, Ukraine; (V.K.); (A.U.); (O.T.)
- Department of Cytology, Bogomoletz Institute of Physiology NAS of Ukraine, 01024 Kyiv, Ukraine
| | - Nataliya V. Bulgakova
- Department of Movement Physiology, Bogomoletz Institute of Physiology NAS of Ukraine, 01024 Kyiv, Ukraine; (N.V.B.); (D.O.Z.)
| | - Danylo O. Zavodovskiy
- Department of Movement Physiology, Bogomoletz Institute of Physiology NAS of Ukraine, 01024 Kyiv, Ukraine; (N.V.B.); (D.O.Z.)
| | - Inna Sokolowska
- Department of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
| | - Andriy Maznychenko
- Department of Movement Physiology, Bogomoletz Institute of Physiology NAS of Ukraine, 01024 Kyiv, Ukraine; (N.V.B.); (D.O.Z.)
- Department of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
- Correspondence: ; Tel.: +38-044-256-24-12
| |
Collapse
|
14
|
Zou T, Jiang S, Yi B, Chen Q, Heng BC, Zhang C. Gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor enhances small molecule-induced neurogenic differentiation of stem cells from apical papilla. J Biomed Mater Res A 2021; 110:623-634. [PMID: 34590393 DOI: 10.1002/jbm.a.37315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
The limited neurogenic potential of adult stem cells and their non-specific lineage differentiation pose major challenges in cell-replacement therapy for neurological disorders. In our previous study, we demonstrated that the neurogenic potential of stem cells from apical papilla (SCAPs) was significantly improved upon induction with a small molecule cocktail. This study attempted to investigate whether neuronal differentiation of SCAPs induced by a small molecule cocktail can be further enhanced in a three-dimensional gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor (BDNF-GelMA). The physiological properties and neural differentiation of SCAPs treated with a combination of small molecules and BDNF-GelMA were evaluated by CCK8, Live/Dead assay, quantitative reverse transcription-polymerase chain reaction, western blot and immunocytochemistry. SCAPs embedded in BDNF-GelMA displayed superior morphological characteristics when induced by a small molecule cocktail, similar to neuronal phenotypes as compared to pure GelMA. There was significant upregulation of neural markers including Tuj1 and MAP2 by SCAPs embedded in BDNF-GelMA, as compared to pure GelMA. Hence, GelMA hydrogel loaded with a potent neurotrophic factor (BDNF) provides a conducive scaffold that can further enhance the differentiation of small molecule-treated SCAPs into neuronal-like cells, which may provide a therapeutic platform for the management of neurological disorders.
Collapse
Affiliation(s)
- Ting Zou
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shan Jiang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Baicheng Yi
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qixin Chen
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | | | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
15
|
Saffari S, Saffari TM, Ulrich DJO, Hovius SER, Shin AY. The interaction of stem cells and vascularity in peripheral nerve regeneration. Neural Regen Res 2021; 16:1510-1517. [PMID: 33433464 PMCID: PMC8323682 DOI: 10.4103/1673-5374.303009] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The degree of nerve regeneration after peripheral nerve injury can be altered by the microenvironment at the site of injury. Stem cells and vascularity are postulated to be a part of a complex pathway that enhances peripheral nerve regeneration; however, their interaction remains unexplored. This review aims to summarize current knowledge on this interaction, including various mechanisms through which trophic factors are promoted by stem cells and angiogenesis. Angiogenesis after nerve injury is stimulated by hypoxia, mediated by vascular endothelial growth factor, resulting in the growth of pre-existing vessels into new areas. Modulation of distinct signaling pathways in stem cells can promote angiogenesis by the secretion of various angiogenic factors. Simultaneously, the importance of stem cells in peripheral nerve regeneration relies on their ability to promote myelin formation and their capacity to be influenced by the microenvironment to differentiate into Schwann-like cells. Stem cells can be acquired through various sources that correlate to their differentiation potential, including embryonic stem cells, neural stem cells, and mesenchymal stem cells. Each source of stem cells serves its particular differentiation potential and properties associated with the promotion of revascularization and nerve regeneration. Exosomes are a subtype of extracellular vesicles released from cell types and play an important role in cell-to-cell communication. Exosomes hold promise for future transplantation applications, as these vesicles contain fewer membrane-bound proteins, resulting in lower immunogenicity. This review presents pre-clinical and clinical studies that focus on selecting the ideal type of stem cell and optimizing stem cell delivery methods for potential translation to clinical practice. Future studies integrating stem cell-based therapies with the promotion of angiogenesis may elucidate the synergistic pathways and ultimately enhance nerve regeneration.
Collapse
Affiliation(s)
- Sara Saffari
- Division of Hand and Microvascular Surgery, Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Plastic and Reconstructive Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tiam M Saffari
- Division of Hand and Microvascular Surgery, Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Plastic and Reconstructive Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dietmar J O Ulrich
- Department of Plastic and Reconstructive Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Steven E R Hovius
- Department of Plastic and Reconstructive Surgery, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alexander Y Shin
- Division of Hand and Microvascular Surgery, Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Vasanthan J, Gurusamy N, Rajasingh S, Sigamani V, Kirankumar S, Thomas EL, Rajasingh J. Role of Human Mesenchymal Stem Cells in Regenerative Therapy. Cells 2020; 10:E54. [PMID: 33396426 PMCID: PMC7823630 DOI: 10.3390/cells10010054] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells which can proliferate and replace dead cells in the body. MSCs also secrete immunomodulatory molecules, creating a regenerative microenvironment that has an excellent potential for tissue regeneration. MSCs can be easily isolated and grown in vitro for various applications. For the past two decades, MSCs have been used in research, and many assays and tests have been developed proving that MSCs are an excellent cell source for therapy. This review focusses on quality control parameters required for applications of MSCs including colony formation, surface markers, differentiation potentials, and telomere length. Further, the specific mechanisms of action of MSCs under various conditions such as trans-differentiation, cell fusion, mitochondrial transfer, and secretion of extracellular vesicles are discussed. This review aims to underline the applications and benefits of MSCs in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
| | - Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Genetic Engineering, SRM Institute of Science and Technology, Chennai 600036, India
| | - Edwin L. Thomas
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (J.V.); (N.G.); (S.R.); (V.S.); (S.K.); (E.L.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
17
|
Juan CH, Chen MH, Lin FH, Wong CS, Chien CC, Chen MH. In Vitro Differentiation of Human Placenta-Derived Multipotent Cells into Schwann-Like Cells. Biomolecules 2020; 10:biom10121657. [PMID: 33322066 PMCID: PMC7763858 DOI: 10.3390/biom10121657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Human placenta-derived multipotent stem cells (PDMCs) resembling embryonic stem cells can differentiate into three germ layer cells, including ectodermal lineage cells, such as neurons, astrocytes, and oligodendrocytes. The favorable characteristics of noninvasive cell harvesting include fewer ethical, religious, and legal considerations as well as accessible and limitless supply. Thus, PDMCs are attractive for cell-based therapy. The Schwann cell (SC) is the most common cell type used for tissue engineering such as nerve regeneration. However, the differentiation potential of human PDMCs into SCs has not been demonstrated until now. In this study, we evaluated the potential of PDMCs to differentiate into SC-like cells in a differentiation medium. After induction, PDMCs not only exhibited typical SC spindle-shaped morphology but also expressed SC markers, including S100, GFAP, p75, MBP, and Sox 10, as revealed by immunocytochemistry. Moreover, a reverse transcription-quantitative polymerase chain reaction analysis revealed the elevated gene expression of S100, GFAP, p75, MBP, Sox-10, and Krox-20 after SC induction. A neuroblastoma cell line, SH-SY5Y, was cultured in the conditioned medium (CM) collected from PDMC-differentiated SCs. The growth rate of the SH-SY5Y increased in the CM, indicating the function of PDMC-induced SCs. In conclusion, human PDMCs can be differentiated into SC-like cells and thus are an attractive alternative to SCs for cell-based therapy in the future.
Collapse
Affiliation(s)
- Chung-Hau Juan
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan; (C.-H.J.); (C.-S.W.); (C.-C.C.)
- Department of Biomedical Sciences, National Central University, Taoyuan 32001, Taiwan
| | - Mei-Hsiu Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
- Department of Biomedical Engineering, Ming Chuan University, Taoyuan 333321, Taiwan
| | - Feng-Hui Lin
- Department of Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan;
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan; (C.-H.J.); (C.-S.W.); (C.-C.C.)
| | - Chih-Cheng Chien
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan; (C.-H.J.); (C.-S.W.); (C.-C.C.)
| | - Ming-Hong Chen
- Department of Neurosurgery, Taipei Municipal Wangfang Hospital, Taipei 116081, Taiwan
- Department of Biomedical Sciences, Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110301, Taiwan
- Correspondence:
| |
Collapse
|
18
|
Choudhary P, Gupta A, Singh S. Therapeutic Advancement in Neuronal Transdifferentiation of Mesenchymal Stromal Cells for Neurological Disorders. J Mol Neurosci 2020; 71:889-901. [PMID: 33047251 DOI: 10.1007/s12031-020-01714-5] [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: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders have become the leading cause of chronic pain and death. Treatments available are not sufficient to help the patients as they only alleviate the symptoms and not the cause. In this regard, stem cells therapy has emerged as an upcoming option for the replacement of dead and damaged neurons. Stem cells, in general, are characterized as cells exhibiting potency properties, i.e., on being subjected to specific conditions they transform into cells of another lineage. Of all the types, mesenchymal stem cells (MSCs) are known for their pluripotent nature without the obstacle of ethical concern surrounding the procurement of other cell types. Although fibroblasts are quite similar to MSCs morphologically, certain markers like CD73, CD 90 are specific to MSCs, making both the cell types distinguishable from each other. This is implemented while procuring MSCs from a plethora of sources like umbilical cord blood, adipose tissue, bone marrow, etc. Among these, bone marrow MSCs are the most widely used type for neural regeneration. Neural regeneration is achieved via transdifferentiation. Several studies have either transplanted the stem cells into rodent models or have carried out transdifferentiation in vitro. The process involves a combination of growth factors, pre-treatment factors, and neuronal differentiation inducing mediums. The results obtained are characterized by neuron-like morphology, expression of markers, along with electrophysical activity in some. Recent attempts involve exploring biomaterials that may mimic the native ECM and therefore can be directly introduced at the site of interest. The review gives a brief description of MSCs, their sources and markers, and the different attempts that have been made towards achieving the goal of differentiating MSCs into neurons.
Collapse
Affiliation(s)
- Princy Choudhary
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Ayushi Gupta
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Sangeeta Singh
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India.
| |
Collapse
|
19
|
Shea GK, Tai EW, Leung KH, Mung AK, Li MT, Tsui AY, Tam AK, Shum DK, Chan Y. Juxtacrine signalling via Notch and ErbB receptors in the switch to fate commitment of bone marrow‐derived Schwann cells. Eur J Neurosci 2020; 52:3306-3321. [DOI: 10.1111/ejn.14837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 05/03/2020] [Accepted: 05/18/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Graham Ka‐Hon Shea
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
| | - Evelyn Wing‐Yin Tai
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Katherine Ho‐Yan Leung
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Alan Kwan‐Long Mung
- Department of Orthopaedics and Traumatology Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong Hong Kong
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
| | - Maximilian Tak‐Sui Li
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Alex Yat‐Ping Tsui
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Anthony Kin‐Wai Tam
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
| | - Daisy Kwok‐Yan Shum
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
- State Key Laboratory of Brain and Cognitive Science The University of Hong Kong Hong Kong Hong Kong
| | - Ying‐Shing Chan
- Li Ka Shing Faculty of Medicine School of Biomedical Sciences The University of Hong Kong Hong Kong Hong Kong
- Research Centre of Heart, Brain, Hormone & Healthy Aging The University of Hong Kong Hong Kong Hong Kong
- State Key Laboratory of Brain and Cognitive Science The University of Hong Kong Hong Kong Hong Kong
| |
Collapse
|
20
|
Ex-Vivo Stimulation of Adipose Stem Cells by Growth Factors and Fibrin-Hydrogel Assisted Delivery Strategies for Treating Nerve Gap-Injuries. Bioengineering (Basel) 2020; 7:bioengineering7020042. [PMID: 32380789 PMCID: PMC7357460 DOI: 10.3390/bioengineering7020042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/22/2022] Open
Abstract
Peripheral nerve injuries often result in lifelong disabilities despite advanced surgical interventions, indicating the urgent clinical need for effective therapies. In order to improve the potency of adipose-derived stem cells (ASC) for nerve regeneration, the present study focused primarily on ex-vivo stimulation of ASC by using growth factors, i.e., nerve growth factor (NGF) or vascular endothelial growth factor (VEGF) and secondly on fibrin-hydrogel nerve conduits (FNC) assisted ASC delivery strategies, i.e., intramural vs. intraluminal loading. ASC were stimulated by NGF or VEGF for 3 days and the resulting secretome was subsequently evaluated in an in vitro axonal outgrowth assay. For the animal study, a 10 mm sciatic nerve gap-injury was created in rats and reconstructed using FNC loaded with ASC. Secretome derived from NGF-stimulated ASC promoted significant axonal outgrowth from the DRG-explants in comparison to all other conditions. Thus, NGF-stimulated ASC were further investigated in animals and found to enhance early nerve regeneration as evidenced by the increased number of β-Tubulin III+ axons. Notably, FNC assisted intramural delivery enabled the improvement of ASC’s therapeutic efficacy in comparison to the intraluminal delivery system. Thus, ex-vivo stimulation of ASC by NGF and FNC assisted intramural delivery may offer new options for developing effective therapies.
Collapse
|
21
|
Zhou LN, Wang JC, Zilundu PLM, Wang YQ, Guo WP, Zhang SX, Luo H, Zhou JH, Deng RD, Chen DF. A comparison of the use of adipose-derived and bone marrow-derived stem cells for peripheral nerve regeneration in vitro and in vivo. Stem Cell Res Ther 2020; 11:153. [PMID: 32272974 PMCID: PMC7147018 DOI: 10.1186/s13287-020-01661-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/15/2020] [Accepted: 03/23/2020] [Indexed: 01/08/2023] Open
Abstract
Background To date, it has repeatedly been demonstrated that infusing bone marrow-derived stem cells (BMSCs) into acellular nerve scaffolds can promote and support axon regeneration through a peripheral nerve defect. However, harvesting BMSCs is an invasive and painful process fraught with a low cellular yield. Methods In pursuit of alternative stem cell sources, we isolated stem cells from the inguinal subcutaneous adipose tissue of adult Sprague–Dawley rats (adipose-derived stem cells, ADSCs). We used a co-culture system that allows isolated adult mesenchymal stem cells (MSCs) and Schwann cells (SCs) to grow in the same culture medium but without direct cellular contact. We verified SC phenotype in vitro by cell marker analysis and used red fluorescent protein-tagged ADSCs to detect their fate after being injected into a chemically extracted acellular nerve allograft (CEANA). To compare the regenerative effects of CEANA containing either BMSCs or ADSCs with an autograft and CEANA only on the sciatic nerve defect in vivo, we performed histological and functional assessments up to 16 weeks after grafting. Results In vitro, we observed reciprocal beneficial effects of ADSCs and SCs in the ADSC–SC co-culture system. Moreover, ADSCs were able to survive in CEANA for 5 days after in vitro implantation. Sixteen weeks after grafting, all results consistently showed that CEANA infused with BMSCs or ADSCs enhanced injured sciatic nerve repair compared to the acellular CEANA-only treatment. Furthermore, their beneficial effects on sciatic injury regeneration were comparable as histological and functional parameters evaluated showed no statistically significant differences. However, the autograft group was roundly superior to both the BMSC- or ADSC-loaded CEANA groups. Conclusion The results of the present study show that ADSCs are a viable alternative stem cell source for treating sciatic nerve injury in lieu of BMSCs.
Collapse
Affiliation(s)
- Li Na Zhou
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China.
| | - Jia Chuan Wang
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | | | - Ya Qiong Wang
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wen Ping Guo
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Sai Xia Zhang
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Hui Luo
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Jian Hong Zhou
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Ru Dong Deng
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China
| | - Dong Feng Chen
- Department of Anatomy, School of basic medical sciences, Guangzhou University of Chinese Medicine, 232 Waihuan East Road, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
22
|
Attia N, Mashal M. Mesenchymal Stem Cells: The Past Present and Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1312:107-129. [PMID: 33159306 DOI: 10.1007/5584_2020_595] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The biomedical applications of mesenchymal stem cells (MSCs) have gained expanding attention over the past three decades. MSCs are easily obtained from various tissue types (e.g. bone marrow, fat, cord blood, etc.), are capable of self-renewal, and could be induced to differentiate into several cell lineages for countless biomedical applications. In addition, when transplanted, MSCs are not detected by immune surveillance, thus do not lead to graft rejection. Moreover, they can home towards affected tissues and induce their therapeutic effect in a cell-base and/or a cell-free manner. These properties, and many others, have made MSCs appealing therapeutic cell candidates (for cell and/or gene therapy) in myriad clinical conditions. However, similar to any other therapeutic tool, MSCs still have their own limitations and grey areas that entail more research for better understanding and optimization. Herein, we present a brief overview of various pre-clinical/clinical applications of MSCs in regenerative medicine and discuss limitations and future challenges.
Collapse
Affiliation(s)
- Noha Attia
- Department of Basic Sciences, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda. .,The Center of research and evaluation, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda. .,Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria, Egypt. .,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
| | - Mohamed Mashal
- The Center of research and evaluation, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda.,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
23
|
Jeong SK, Choi I, Jeon SR. Current Status and Future Strategies to Treat Spinal Cord Injury with Adult Stem Cells. J Korean Neurosurg Soc 2019; 63:153-162. [PMID: 31805758 PMCID: PMC7054109 DOI: 10.3340/jkns.2019.0146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) is one of the most devastating conditions and many SCI patients suffer neurological sequelae. Stem cell therapies are expected to be beneficial for many patients with central nervous system injuries, including SCI. Adult stem cells (ASCs) are not associated with the risks which embryonic stem cells have such as malignant transformation, or ethical problems, and can be obtained relatively easily. Consequently, many researchers are currently studying the effects of ASCs in clinical trials. The environment of transplanted cells applied in the injured spinal cord differs between the phases of SCI; therefore, many researchers have investigated these phases to determine the optimal time window for stem cell therapy in animals. In addition, the results of clinical trials should be evaluated according to the phase in which stem cells are transplanted. In general, the subacute phase is considered to be optimal for stem cell transplantation. Among various candidates of transplantable ASCs, mesenchymal stem cells (MSCs) are most widely studied due to their clinical safety. MSCs are also less immunogenic than neural stem/progenitor cells and consequently immunosuppressants are rarely required. Attempts have been made to enhance the effects of stem cells using scaffolds, trophic factors, cytokines, and other drugs in animal and/or human clinical studies. Over the past decade, several clinical trials have suggested that transplantation of MSCs into the injured spinal cord elicits therapeutic effects on SCI and is safe; however, the clinical effects are limited at present. Therefore, new therapeutic agents, such as genetically enhanced stem cells which effectively secrete neurotrophic factors or cytokines, must be developed based on the safety of pure MSCs.
Collapse
Affiliation(s)
- Seong Kyun Jeong
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Il Choi
- Department of Neurological Surgery, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Sang Ryong Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
24
|
Manoukian OS, Baker JT, Rudraiah S, Arul MR, Vella AT, Domb AJ, Kumbar SG. Functional polymeric nerve guidance conduits and drug delivery strategies for peripheral nerve repair and regeneration. J Control Release 2019; 317:78-95. [PMID: 31756394 DOI: 10.1016/j.jconrel.2019.11.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/16/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022]
Abstract
Peripheral nerve injuries can be extremely debilitating, resulting in sensory and motor loss-of-function. Endogenous repair is limited to non-severe injuries in which transection of nerves necessitates surgical intervention. Traditional treatment approaches include the use of biological grafts and alternative engineering approaches have made progress. The current article serves as a comprehensive, in-depth perspective on peripheral nerve regeneration, particularly nerve guidance conduits and drug delivery strategies. A detailed background of peripheral nerve injury and repair pathology, and an in-depth look into augmented nerve regeneration, nerve guidance conduits, and drug delivery strategies provide a state-of-the-art perspective on the field.
Collapse
Affiliation(s)
- Ohan S Manoukian
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA; Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Jiana T Baker
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Swetha Rudraiah
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA; Department of Pharmaceutical Sciences, University of Saint Joseph, Hartford, CT, USA
| | - Michael R Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Anthony T Vella
- Department of Department of Immunology, University of Connecticut Health, Farmington, CT, USA
| | - Abraham J Domb
- Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Sangamesh G Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA; Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA.
| |
Collapse
|
25
|
Yang Q, Lopez MJ. The Equine Hoof: Laminitis, Progenitor (Stem) Cells, and Therapy Development. Toxicol Pathol 2019; 49:1294-1307. [PMID: 31741428 DOI: 10.1177/0192623319880469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The equine hoof capsule, composed of modified epidermis and dermis, is vital for protecting the third phalanx from forces of locomotion. There are descriptions of laminitis, defined as inflammation of sensitive hoof tissues but recognized as pathologic changes with or without inflammatory mediators, in the earliest records of domesticated horses. Laminitis can range from mild to serious, and signs can be acute, chronic, or transition from acute, severe inflammation to permanently abnormal tissue. Damage within the intricate dermal and epidermal connections of the primary and secondary lamellae is often associated with lifelong changes in hoof growth, repair, and conformation. Decades of research contribute to contemporary standards of care that include systemic and local therapies as well as mechanical hoof support. Despite this, consistent mechanisms to restore healthy tissue formation following a laminitic insult are lacking. Endogenous and exogenous progenitor cell contributions to healthy tissue formation is established for most tissues. There is comparably little information about equine hoof progenitor cells. Equine hoof anatomy, laminitis, and progenitor cells are covered in this review. The potential of progenitor cells to advance in vitro equine hoof tissue models and translate to clinical therapies may significantly improve prevention and treatment of a devastating condition that has afflicted equine companions throughout history.
Collapse
Affiliation(s)
- Qingqiu Yang
- Department of Veterinary Clinical Sciences, Laboratory for Equine and Comparative Orthopedic Research, Baton Rouge, LA, USA
| | - Mandi J Lopez
- Department of Veterinary Clinical Sciences, Laboratory for Equine and Comparative Orthopedic Research, Baton Rouge, LA, USA
| |
Collapse
|
26
|
Manoukian OS, Stratton S, Arul MR, Moskow J, Sardashti N, Yu X, Rudraiah S, Kumbar SG. Polymeric ionically conductive composite matrices and electrical stimulation strategies for nerve regeneration: In vitro characterization. J Biomed Mater Res B Appl Biomater 2019; 107:1792-1805. [PMID: 30419159 PMCID: PMC6511498 DOI: 10.1002/jbm.b.34272] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/18/2018] [Accepted: 07/21/2018] [Indexed: 12/14/2022]
Abstract
Stem cell strategies and the use of electrical stimulation (ES) represent promising new frontiers for peripheral nerve regeneration. Composite matrices were fabricated by coating electrospun polycaprolactone/cellulose acetate micro-nanofibers with chitosan and ionically conductive (IC) polymers including, sulfonated polyaniline, and lignin sulfonate. These composite matrices were characterized for surface morphology, coating uniformity, ionic conductivity, and mechanical strength to explore as scaffold materials for nerve regeneration in conjunction with ES. Composite matrices measured conductivity in the range of 0.0049-0.0068 mS/m due to the uniform coating of sulfonated polymers on the micro-nanofibers. Thin films (2D) and composite fiber matrices (3D) of IC polymers seeded with human mesenchymal stem cells (hMSCs) were electrically stimulated at 0.5 V, 20 Hz for 1 h daily for 14 days to study the changes in cell viability, morphology, and expression of the neuronal-like phenotype. In vitro ES lead to changes in hMSCs' fibroblast morphology into elongated neurite-like structures with cell bodies for ES-treated and positive control growth factor-treated groups. Immunofluorescent staining revealed the presence of neuronal markers including β3-tubulin, microtubule-associated protein 2, and nestin in response to ES. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1792-1805, 2019.
Collapse
Affiliation(s)
- Ohan S. Manoukian
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Scott Stratton
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Michael R. Arul
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Joshua Moskow
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Naseem Sardashti
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Xiaojun Yu
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, USA
| | - Swetha Rudraiah
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
- Department of Pharmaceutical Sciences, University of Saint Joseph, Hartford, CT, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
27
|
Endo T, Kadoya K, Suzuki Y, Kawamura D, Iwasaki N. A Novel Experimental Model to Determine the Axon-Promoting Effects of Grafted Cells After Peripheral Nerve Injury. Front Cell Neurosci 2019; 13:280. [PMID: 31316351 PMCID: PMC6611175 DOI: 10.3389/fncel.2019.00280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022] Open
Abstract
Although peripheral nerves can regenerate, clinical outcomes after peripheral nerve injuries are not always satisfactory, especially in cases of severe or proximal injuries. Further, autologous nerve grafting remains the gold standard for the reconstruction of peripheral nerves, although this method is still accompanied by issues of donor-site morbidity and limited supply. Cell therapy is a potential approach to overcome these issues. However, the optimal cell type for promoting axon regeneration remains unknown. Here, we report a novel experimental model dedicated to elucidation of the axon-promoting effects of candidate cell types using simple and standardized techniques. This model uses rat sciatic nerves and consists of a 25 mm-long acellular region and a crush site at each end. The acellular region was made by repeated freeze/thaw procedures with liquid nitrogen. Importantly, the new model does not require microsurgical procedures, which are technically demanding and greatly affect axon regeneration. To test the actual utility of this model, red fluorescent protein-expressing syngeneic Schwann cells (SCs), marrow stromal cells, or fibroblasts were grafted into the acellular area, followed by perfusion of the rat 2 weeks later. All types of grafted cells survived well. Quantification of regenerating axons demonstrated that SCs, but not the other cell types, promoted axon regeneration with minimum variability. Thus, this model is useful for differentiating the effects of various grafted cell types in axon regeneration. Interestingly, regardless of the grafted cell type, host SCs migrated into the acellular area, and the extent of axon regeneration was strongly correlated with the number of SCs. Moreover, all regenerating axons were closely associated with SCs. These findings suggest a critical role for SCs in peripheral nerve axon regeneration. Collectively, this novel experimental model is useful for elucidating the axon-promoting effects of grafted cells and for analyzing the biology of peripheral nerve axon regeneration.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuki Suzuki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daisuke Kawamura
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
28
|
Bassilios Habre S, Bond G, Jing XL, Kostopoulos E, Wallace RD, Konofaos P. The Surgical Management of Nerve Gaps: Present and Future. Ann Plast Surg 2019; 80:252-261. [PMID: 29166306 DOI: 10.1097/sap.0000000000001252] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peripheral nerve injuries can result in significant morbidity, including motor and/or sensory loss, which can affect significantly the life of the patient. Nowadays, the gold standard for the treatment of nerve section is end-to-end neurorrhaphy. Unfortunately, in some cases, there is segmental loss of the nerve trunk. Nerve mobilization allows primary repair of the sectioned nerve by end-to-end neurorrhaphy if the gap is less than 1 cm. When the nerve gap exceeds 1 cm, autologous nerve grafting is the gold standard of treatment. To overcome the limited availability and the donor site morbidity, other techniques have been used: vascularized nerve grafts, cellular and acellular allografts, nerve conduits, nerve transfers, and end-to-side neurorrhaphy. The purpose of this review is to present an overview of the literature on the applications of these techniques in peripheral nerve repair. Furthermore, preoperative evaluation, timing of repair, and future perspectives are also discussed.
Collapse
|
29
|
Rbia N, Bulstra LF, Lewallen EA, Hovius SER, van Wijnen AJ, Shin AY. Seeding decellularized nerve allografts with adipose-derived mesenchymal stromal cells: An in vitro analysis of the gene expression and growth factors produced. J Plast Reconstr Aesthet Surg 2019; 72:1316-1325. [PMID: 31175032 DOI: 10.1016/j.bjps.2019.04.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 02/07/2019] [Accepted: 04/27/2019] [Indexed: 12/24/2022]
Abstract
Mesenchymal stromal cells (MSCs) secrete many soluble growth factors and have previously been shown to stimulate nerve regeneration. MSC-seeded processed nerve allografts could potentially be a promising method for large segmental motor nerve injuries. Further progress in our understanding of how the functions of MSCs can be leveraged for peripheral nerve repair is required before making clinical translation. The present study, therefore, investigated whether interactions of adipose-derived MSCs with decellularized nerve allografts can improve gene and protein expression of growth factors that may support nerve regeneration. Human nerve allografts (n = 30) were decellularized and seeded with undifferentiated human adipose-derived MSCs. Subsequently, the MSCs and MSC-seeded grafts were isolated on days 3, 7, 14, and 21 in culture for RNA expression analysis by qRT-PCR. Evaluated genes included NGF, BDNF, PTN, GAP43, MBP, PMP22, VEGF, and CD31. Growth factor production was evaluated and quantified using enzyme-linked immunosorbent assay (ELISA). On day 21, semi-quantitative RT-PCR analysis showed that adherence of MSCs to nerve allografts significantly enhances mRNA expression of neurotrophic, angiogenic, endothelial, and myelination markers (e.g., BDNF, VEGF, CD31, and MBP). ELISA results revealed an upregulation of BDNF and reduction of both VEGF and NGF protein levels. This study demonstrates that seeding of undifferentiated adipose-derived MSCs onto processed nerve allografts permits the secretion of neurotrophic and angiogenic factors that can stimulate nerve regeneration. These favorable molecular changes suggest that MSC supplementation of nerve allografts may have potential in improving nerve regeneration.
Collapse
Affiliation(s)
- Nadia Rbia
- Department of Orthopedic Surgery, Division of Hand and Microvascular Surgery, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905, USA; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, 's Gravendijkwal 230, 3015 CE Rotterdam, the Netherlands
| | - Liselotte F Bulstra
- Department of Orthopedic Surgery, Division of Hand and Microvascular Surgery, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905, USA; Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, 's Gravendijkwal 230, 3015 CE Rotterdam, the Netherlands
| | - Eric A Lewallen
- Department of Orthopedic Surgery, Division of Hand and Microvascular Surgery, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905, USA; Department of Biological Sciences, Hampton University, 100 E Queen St, Hampton, VA 23668, USA
| | - Steven E R Hovius
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus Medical Center, 's Gravendijkwal 230, 3015 CE Rotterdam, the Netherlands; Xpert Clinic, Hand and Wrist Surgery, Jan Leentvaarlaan 14-24, 3065 DC Rotterdam, the Netherlands
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Division of Hand and Microvascular Surgery, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905, USA
| | - Alexander Y Shin
- Department of Orthopedic Surgery, Division of Hand and Microvascular Surgery, Mayo Clinic, 200 First Street S.W., Rochester, MN 55905, USA.
| |
Collapse
|
30
|
Rink S, Bendella H, Akkin SM, Manthou M, Grosheva M, Angelov DN. Experimental Studies on Facial Nerve Regeneration. Anat Rec (Hoboken) 2019; 302:1287-1303. [DOI: 10.1002/ar.24123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/09/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral MedicineUniversity of Cologne Cologne Germany
| | - Habib Bendella
- Department of NeurosurgeryUniversity of Witten/Herdecke, Cologne Merheim Medical Center (CMMC) Cologne Germany
| | - Salih Murat Akkin
- Department of Anatomy, School of MedicineSANKO University Gaziantep Turkey
| | - Marilena Manthou
- Department of Histology and EmbryologyAristotle University Thessaloniki Thessaloniki Greece
| | - Maria Grosheva
- Department of Oto‐Rhino‐LaryngologyUniversity of Cologne Cologne Germany
| | | |
Collapse
|
31
|
Boecker A, Daeschler SC, Kneser U, Harhaus L. Relevance and Recent Developments of Chitosan in Peripheral Nerve Surgery. Front Cell Neurosci 2019; 13:104. [PMID: 31019452 PMCID: PMC6458244 DOI: 10.3389/fncel.2019.00104] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 02/28/2019] [Indexed: 12/20/2022] Open
Abstract
Developments in tissue engineering yield biomaterials with different supporting strategies to promote nerve regeneration. One promising material is the naturally occurring chitin derivate chitosan. Chitosan has become increasingly important in various tissue engineering approaches for peripheral nerve reconstruction, as it has demonstrated its potential to interact with regeneration associated cells and the neural microenvironment, leading to improved axonal regeneration and less neuroma formation. Moreover, the physiological properties of its polysaccharide structure provide safe biodegradation behavior in the absence of negative side effects or toxic metabolites. Beneficial interactions with Schwann cells (SC), inducing differentiation of mesenchymal stromal cells to SC-like cells or creating supportive conditions during axonal recovery are only a small part of the effects of chitosan. As a result, an extensive body of literature addresses a variety of experimental strategies for the different types of nerve lesions. The different concepts include chitosan nanofibers, hydrogels, hollow nerve tubes, nerve conduits with an inner chitosan layer as well as hybrid architectures containing collagen or polyglycolic acid nerve conduits. Furthermore, various cell seeding concepts have been introduced in the preclinical setting. First translational concepts with hollow tubes following nerve surgery already transferred the promising experimental approach into clinical practice. However, conclusive analyses of the available data and the proposed impact on the recovery process following nerve surgery are currently lacking. This review aims to give an overview on the physiologic properties of chitosan, to evaluate its effect on peripheral nerve regeneration and discuss the future translation into clinical practice.
Collapse
Affiliation(s)
- A Boecker
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - S C Daeschler
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - U Kneser
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - L Harhaus
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| |
Collapse
|
32
|
Urrutia DN, Caviedes P, Mardones R, Minguell JJ, Vega-Letter AM, Jofre CM. Comparative study of the neural differentiation capacity of mesenchymal stromal cells from different tissue sources: An approach for their use in neural regeneration therapies. PLoS One 2019; 14:e0213032. [PMID: 30856179 PMCID: PMC6437714 DOI: 10.1371/journal.pone.0213032] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/13/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can trans/differentiate to neural precursors and/or
mature neurons and promote neuroprotection and neurogenesis. The above could
greatly benefit neurodegenerative disorders as well as in the treatment of
post-traumatic and hereditary diseases of the central nervous system (CNS). In
order to attain an ideal source of adult MSCs for the treatment of CNS diseases,
adipose tissue, bone marrow, skin and umbilical cord derived MSCs were isolated
and studied to explore differences with regard to neural differentiation
capacity. In this study, we demonstrated that MSCs from several tissues can
differentiate into neuron-like cells and differentially express progenitors and
mature neural markers. Adipose tissue MSCs exhibited significantly higher
expression of neural markers and had a faster proliferation rate. Our results
suggest that adipose tissue MSCs are the best candidates for the use in
neurological diseases.
Collapse
Affiliation(s)
| | - Pablo Caviedes
- Program of Molecular & Clinical Pharmacology, ICBM, Faculty of
Medicine, Universidad de Chile, Santiago, Chile
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de
Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y
Matemáticas, Universidad de Chile, Santiago, Chile
| | - Rodrigo Mardones
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- Orthopedic Department, Clinica Las Condes, Santiago,
Chile
| | - José J. Minguell
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
| | - Ana Maria Vega-Letter
- Program of Traslational Immunology ICIM, Faculty of Medicine, Clinica
Alemana Universidad del Desarrollo, Santiago, Chile
| | - Claudio M. Jofre
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- * E-mail:
| |
Collapse
|
33
|
The interaction between SBA-15 derivative loaded with Ph 3Sn(CH 2) 6OH and human melanoma A375 cell line: uptake and stem phenotype loss. J Biol Inorg Chem 2019; 24:223-234. [PMID: 30759278 DOI: 10.1007/s00775-019-01640-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/21/2018] [Indexed: 12/13/2022]
Abstract
Extraordinary progress in medicinal inorganic chemistry in the past few years led to the rational design of novel platinum compounds, as well as nonplatinum metal-based antitumor agents, including organotin compounds, whose activity is not based on unrepairable interaction with DNA. To overcome poor solubility and toxicity problems that limited the application of these compounds numerous delivering systems were used (Lila et al. in Biol Pharm Bull 37:206-211, 2014; Yue and Cao in Curr Cancer Drug Targets 16:480-488, 2016; Duan et al. in WIREs Nanomed Nanobiotechnol 8:776-791, 2016). Regarding high drug loading capacity, mesoporous silica nanoparticles like SBA-15 became more important for targeted drug delivery. In this study, cellular uptake and biological activities responsible for organotin(IV) compound Ph3Sn(CH2)6OH (Sn6) grafted into (3-chloropropyl)triethoxysilane functionalized SBA-15 (SBA-15p → SBA-15p|Sn6) were evaluated in human melanoma A375 cell line. Moreover, the influence of SBA-15p grafted with organotin(IV) compound on the stemness of A375 cell was tested. Given the fact that SBA-15p|Sn6 nanoparticles are nonspherical and relatively large, their internalization efficiently started even after 15 min with stable adhesion to the cell membrane. After only 2 h of incubation of A375 cells with SBA-15p|Sn6 passive fluid-phase uptake and macropinocytosis were observed. Inside of the cell, treatment with SBA-15p loaded with Sn6 promoted caspase-dependent apoptosis in parallel with senescence development. The subpopulation of cells expressing Schwann-like phenotype arose upon the treatment, while the signaling pathway responsible for maintenance of pluripotency and invasiveness, Wnt, Notch1, and Oct3/4 were modulated towards less aggressive signature. In summary, SBA-15p enhances the efficacy of free Sn6 compound through efficient uptake and well profiled intracellular response followed with decreased stem characteristics of highly invasive A375 melanoma cells.
Collapse
|
34
|
Ramli K, Aminath Gasim I, Ahmad AA, Hassan S, Law ZK, Tan GC, Baharuddin A, Naicker AS, Htwe O, Mohammed Haflah NH, B H Idrus R, Abdullah S, Ng MH. Human bone marrow-derived MSCs spontaneously express specific Schwann cell markers. Cell Biol Int 2019; 43:233-252. [PMID: 30362196 DOI: 10.1002/cbin.11067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
In peripheral nerve injuries, Schwann cells (SC) play pivotal roles in regenerating damaged nerve. However, the use of SC in clinical cell-based therapy is hampered due to its limited availability. In this study, we aim to evaluate the effectiveness of using an established induction protocol for human bone marrow derived-MSC (hBM-MSCs) transdifferentiation into a SC lineage. A relatively homogenous culture of hBM-MSCs was first established after serial passaging (P3), with profiles conforming to the minimal criteria set by International Society for Cellular Therapy (ISCT). The cultures (n = 3) were then subjected to a series of induction media containing β-mercaptoethanol, retinoic acid, and growth factors. Quantitative RT-PCR, flow cytometry, and immunocytochemistry analyses were performed to quantify the expression of specific SC markers, that is, S100, GFAP, MPZ and p75 NGFR, in both undifferentiated and transdifferentiated hBM-MSCs. Based on these analyses, all markers were expressed in undifferentiated hBM-MSCs and MPZ expression (mRNA transcripts) was consistently detected before and after transdifferentiation across all samples. There was upregulation at the transcript level of more than twofolds for NGF, MPB, GDNF, p75 NGFR post-transdifferentiation. This study highlights the existence of spontaneous expression of specific SC markers in cultured hBM-MSCs, inter-donor variability and that MSC transdifferentiation is a heterogenous process. These findings strongly oppose the use of a single marker to indicate SC fate. The heterogenous nature of MSC may influence the efficiency of SC transdifferentiation protocols. Therefore, there is an urgent need to re-define the MSC subpopulations and revise the minimal criteria for MSC identification.
Collapse
Affiliation(s)
- Khairunnisa Ramli
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ifasha Aminath Gasim
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amir Adham Ahmad
- Department of Orthopaedics, School of Medicine, International Medical University, Negeri Sembilan, Malaysia
| | - Shariful Hassan
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Zhe Kang Law
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Azmi Baharuddin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amaramalar Selvi Naicker
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ohnmar Htwe
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Hazla Mohammed Haflah
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ruszymah B H Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shalimar Abdullah
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
35
|
Rodríguez Sánchez DN, de Lima Resende LA, Boff Araujo Pinto G, de Carvalho Bovolato AL, Possebon FS, Deffune E, Amorim RM. Canine Adipose-Derived Mesenchymal Stromal Cells Enhance Neuroregeneration in a Rat Model of Sciatic Nerve Crush Injury. Cell Transplant 2019; 28:47-54. [PMID: 30369261 PMCID: PMC6322136 DOI: 10.1177/0963689718809045] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/27/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Crush injuries in peripheral nerves are frequent and induce long-term disability with motor and sensory deficits. Due to axonal and myelin sheath disruptions, strategies for optimized axonal regeneration are needed. Multipotent mesenchymal stromal cells (MSC) are promising because of their anti-inflammatory properties and secretion of neurotrophins. The present study investigated the effect of canine adipose tissue MSC (Ad-MSC) transplantation in an experimental sciatic nerve crush injury. Wistar rats were divided into three groups: sham ( n = 8); Crush+PBS ( n = 8); Crush+MSC ( n = 8). Measurements of sciatic nerve functional index (SFI), muscle mass, and electromyography (EMG) were performed. Canine Ad-MSC showed mesodermal characteristics (CD34-, CD45-, CD44+, CD90+ and CD105+) and multipotentiality due to chondrogenic, adipogenic, and osteogenic differentiation. SFI during weeks 3 and 4 was significantly higher in the Crush+MSC group ( p < 0.001). During week 4, the EMG latency in the Crush+MSC groups had better near normality ( p < 0.05). The EMG amplitude showed results close to normality during week 4 in the Crush+MSC group ( p < 0.04). There were no statistical differences in muscle weight between the groups ( p > 0.05), but there was a tendency toward weight gain in the Crush+MSC groups. Better motor functional recovery after crush and perineural canine Ad-MSC transplantation was observed during week 2. This was maintained till week 4. In conclusion, the canine Ad-MSC transplantation showed early pro-regenerative effects between 2-4 weeks in the rat model of sciatic nerve crush injury.
Collapse
Affiliation(s)
- Diego Noé Rodríguez Sánchez
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Luiz Antonio de Lima Resende
- Department of Neurology and Psychiatry, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Giovana Boff Araujo Pinto
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Ana Lívia de Carvalho Bovolato
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Fábio Sossai Possebon
- Department of Veterinary Hygiene and Public Health, College of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
| | - Elenice Deffune
- Blood Transfusion Center, Cell Engineering Laboratory, Botucatu Medical School, São Paulo State University (UNESP), São Paulo, Brazil
| | - Rogério Martins Amorim
- Department of Veterinary Clinics, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), São Paulo, Brazil
| |
Collapse
|
36
|
Yousefi F, Lavi Arab F, Saeidi K, Amiri H, Mahmoudi M. Various strategies to improve efficacy of stem cell transplantation in multiple sclerosis: Focus on mesenchymal stem cells and neuroprotection. J Neuroimmunol 2018; 328:20-34. [PMID: 30557687 DOI: 10.1016/j.jneuroim.2018.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 11/30/2018] [Indexed: 02/09/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) which predominantly affect young adults and undergo heavy socioeconomic burdens. Conventional therapeutic modalities for MS mostly downregulate aggressive immune responses and are almost insufficient for management of progressive course of the disease. Mesenchymal stem cells (MSCs), due to both immunomodulatory and neuroprotective properties have been known as practical cells for treatment of neurodegenerative diseases like MS. However, clinical translation of MSCs is associated with some limitations such as short-life engraftment duration, little in vivo trans-differentiation and restricted accessibility into damaged sites. Therefore, laboratory manipulation of MSCs can improve efficacy of MSCs transplantation in MS patients. In this review, we discuss several novel approaches, which can potentially enhance MSCs capabilities for treating MS.
Collapse
Affiliation(s)
- Forouzan Yousefi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kolsoum Saeidi
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Houshang Amiri
- Neurology Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Hou B, Ye Z, Ji W, Cai M, Ling C, Chen C, Guo Y. Comparison of the Effects of BMSC-derived Schwann Cells and Autologous Schwann Cells on Remyelination Using a Rat Sciatic Nerve Defect Model. Int J Biol Sci 2018; 14:1910-1922. [PMID: 30443194 PMCID: PMC6231219 DOI: 10.7150/ijbs.26765] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 09/08/2018] [Indexed: 12/13/2022] Open
Abstract
Schwann cells (SCs) are primarily responsible for the formation of myelin sheaths, yet bone marrow mesenchymal stem cell (BMSC)-derived SCs are often used to replace autologous SCs and assist with the repair of peripheral nerve myelin sheaths. In this study, the effects of the two cell types on remyelination were compared during the repair of peripheral nerves. Methods: An acellular nerve scaffold was prepared using the extraction technique. Rat BMSCs and autologous SCs were extracted. BMSCs were induced to differentiate into BMSC-derived SCs (B-dSCs) in vitro. Seed cells (BMSCs, B-dSCs, and autologous SCs) were cocultured with nerve scaffolds (Sca) in vitro. Rats with severed sciatic nerves were used as the animal model. A composite scaffold was used to bridge the broken ends. After surgery, electrophysiology, cell tracking analyses (EdU labeling), immunofluorescence staining (myelin basic protein (MBP)), toluidine blue staining, and transmission electron microscopy were conducted to compare remyelination between the various groups and to evaluate the effects of the seed cells on myelination. One week after transplantation, only a small number of B-dSCs expressed MBP, which was far less than the proportion of MBP-expressing autologous SCs (P<0.01) but was higher than the proportion of BMSCs expressing MBP (P<0.05). Four weeks after surgery, the electrophysiology results (latency time, conductive velocity and amplitude) and various quantitative indicators of remyelination (thickness, distribution, and the number of myelinated fibers) showed that the Sca+B-dSC group was inferior to the Sca+autologous SC group (P<0.05) but was superior to the Sca+BMSC group (P<0.05). Conclusions: Within 4 weeks after surgery, the use of an acellular nerve scaffold combined with B-dSCs promotes remyelination to a certain extent, but the effect is significantly less than that of the scaffold combined with autologous SCs.
Collapse
Affiliation(s)
- Bo Hou
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Zhuopeng Ye
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Wanqing Ji
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, 510623, China
| | - Meiqin Cai
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| |
Collapse
|
38
|
Galhom RA, Hussein Abd El Raouf HH, Mohammed Ali MH. Role of bone marrow derived mesenchymal stromal cells and Schwann-like cells transplantation on spinal cord injury in adult male albino rats. Biomed Pharmacother 2018; 108:1365-1375. [PMID: 30372839 DOI: 10.1016/j.biopha.2018.09.131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Spinal cord injury is a considerable health impact accompanied with physical, psychological and economic burden. Bone marrow derived mesenchymal stromal cells (BM-MSCs) transplantation was found to produce neuronal regenerative effects. Schwann-like cells differentiated from BM-MSCs have myelin-forming ability. AIM OF THE WORK To compare the ability of BM-MSCs versus Schwann like cells to promote recovery of spinal cord injury. MATERIAL AND METHODS Adult male albino rats were used throughout the study. BM-MSCs were harvested from femora of rats. Sciatic nerves were extracted and used in the preparation of the induction culture medium for differentiation of BM-MSCs into Schwann-like cells. Rats were divided into control, spinal cord injured (SCI), spinal cord injured plus BM-MSCs transplantation (BM-MSC) and spinal cord injured plus Schwann-like cells transplantation (Sn) groups. BBB scale assessment was performed before and after SCI in all rats. Rats were euthanized at the end of the 7th week and spinal cords were dissected and processed for light and transmission electron microscopic examinations. RESULTS Spinal cord sections of SCI group revealed cavitation, necrosis and demyelination. BM-MSC and Sn groups showed both functional and structural improvement compared to SCI group with better BBB score and histopathological features in the BM-MSC group and more expression of S100 in the Sn group. CONCLUSION Transplantation of BM-MSCs and Schwann-like cells improved the structural and functional alterations of spinal cord injury with better improvement in BM-MSC group.
Collapse
Affiliation(s)
- Rania A Galhom
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | | | - Mona H Mohammed Ali
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
39
|
Boecker AH, Bozkurt A, Kim BS, Altinova H, Tank J, Deumens R, Tolba R, Weis J, Brook GA, Pallua N, van Neerven SGA. Cell-enrichment with olfactory ensheathing cells has limited local extra beneficial effects on nerve regeneration supported by the nerve guide Perimaix. J Tissue Eng Regen Med 2018; 12:2125-2137. [PMID: 30044547 DOI: 10.1002/term.2731] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/21/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022]
Abstract
The reconstruction of peripheral nerve injuries is clinically challenging, and today, the autologous nerve transplantation is still considered as the only gold standard remedy for nerve lesions where a direct nerve coaptation is not possible. Nevertheless, the functional merits of many biomaterials have been tested as potential substitutes for the autologous nerve transplant. One of the strategies that have been pursued is the combination of bioengineered nerve guides with cellular enrichment. In this present study, we combined the previously evaluated collagen-based and microstructured nerve guide Perimaix with olfactory ensheathing cell enrichment. Rat sciatic nerve defects of 20 mm were either bridged by a cell-seeded or nonseeded nerve guide or an autologous nerve transplant. Animals were monitored for 12 weeks for structural and functional parameters. Seeded cells survived on Perimaix, and following implantation aligned along the microstructured Perimaix framework. Axonal densities within the cell-seeded nerve guides were higher than in the nonseeded nerve guides and were comparable to the autograft. Additionally, cell-seeding had local beneficial effects on myelination within the nerve guide, as myelin sheath thickness was enhanced when compared with the empty scaffold. Nevertheless, for bridging the nerve gap of 20 mm, both the cell-seeded as well as nonseeded scaffolds were equally efficient regarding the functional outcome, which did not differ between the autograft, seeded or nonseeded groups. Our data demonstrate that olfactory ensheathing cell enrichment has local effects on nerve regeneration in combination with the Perimaix nerve guide. Surprisingly, for traversing the lesion gap, additional cell-seeding is not crucial.
Collapse
Affiliation(s)
- Arne Hendrik Boecker
- Department of Plastic Surgery, Reconstructive and Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany.,Department of Hand-Plastic and Reconstructive Surgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Ahmet Bozkurt
- Department of Plastic Surgery, Reconstructive and Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany.,Department of Plastic, Hand, Reconstructive and Aesthetic Surgery, Helios Klinikum Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | - Bong Sung Kim
- Department of Plastic Surgery, Reconstructive and Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | - Haktan Altinova
- Department of Neurosurgery, University Hospital RWTH Aachen, Aachen, Germany.,Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Julian Tank
- Department of General, Visceral and Thoracic Surgery, Private Medical University, Nuremberg, Germany
| | - Ronald Deumens
- Neuropharmacology, Université Catholique de Louvain, Brussels, Belgium.,Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rene Tolba
- Institute for Laboratory Animal Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany.,Translational Brain Medicine (JARA Brain), Juelich-Aachen Research Alliance, Germany
| | - Gary Anthony Brook
- Institute of Neuropathology, University Hospital RWTH Aachen, Aachen, Germany.,Translational Brain Medicine (JARA Brain), Juelich-Aachen Research Alliance, Germany
| | - Norbert Pallua
- Department of Plastic Surgery, Reconstructive and Hand Surgery, Burn Center, University Hospital RWTH Aachen, Aachen, Germany
| | | |
Collapse
|
40
|
Uz M, Das SR, Ding S, Sakaguchi DS, Claussen JC, Mallapragada SK. Advances in Controlling Differentiation of Adult Stem Cells for Peripheral Nerve Regeneration. Adv Healthc Mater 2018; 7:e1701046. [PMID: 29656561 DOI: 10.1002/adhm.201701046] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 01/08/2018] [Indexed: 01/01/2023]
Abstract
Adult stems cells, possessing the ability to grow, migrate, proliferate, and transdifferentiate into various specific phenotypes, constitute a great asset for peripheral nerve regeneration. Adult stem cells' ability to undergo transdifferentiation is sensitive to various cell-to-cell interactions and external stimuli involving interactions with physical, mechanical, and chemical cues within their microenvironment. Various studies have employed different techniques for transdifferentiating adult stem cells from distinct sources into specific lineages (e.g., glial cells and neurons). These techniques include chemical and/or electrical induction as well as cell-to-cell interactions via co-culture along with the use of various 3D conduit/scaffold designs. Such scaffolds consist of unique materials that possess controllable physical/mechanical properties mimicking cells' natural extracellular matrix. However, current limitations regarding non-scalable transdifferentiation protocols, fate commitment of transdifferentiated stem cells, and conduit/scaffold design have required new strategies for effective stem cells transdifferentiation and implantation. In this progress report, a comprehensive review of recent advances in the transdifferentiation of adult stem cells via different approaches along with multifunctional conduit/scaffolds designs is presented for peripheral nerve regeneration. Potential cellular mechanisms and signaling pathways associated with differentiation are also included. The discussion with current challenges in the field and an outlook toward future research directions is concluded.
Collapse
Affiliation(s)
- Metin Uz
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
| | - Suprem R. Das
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
- Division of Materials Science and Engineering Ames Laboratory Ames IA 50011 USA
| | - Shaowei Ding
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Donald S. Sakaguchi
- Neuroscience Program Iowa State University Ames IA 50011 USA
- Department of Genetics Development and Cell Biology Iowa State University Ames IA 50011 USA
| | - Jonathan C. Claussen
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
- Division of Materials Science and Engineering Ames Laboratory Ames IA 50011 USA
| | - Surya K. Mallapragada
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
- Department of Genetics Development and Cell Biology Iowa State University Ames IA 50011 USA
| |
Collapse
|
41
|
Him A, Onger ME, Delibas B. Periferik Sinir Rejenerasyonu ve Kök Hücre Tedavileri. ACTA ACUST UNITED AC 2018. [DOI: 10.31832/smj.404819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Sun X, Zhu Y, Yin HY, Guo ZY, Xu F, Xiao B, Jiang WL, Guo WM, Meng HY, Lu SB, Wang Y, Peng J. Differentiation of adipose-derived stem cells into Schwann cell-like cells through intermittent induction: potential advantage of cellular transient memory function. Stem Cell Res Ther 2018; 9:133. [PMID: 29751848 PMCID: PMC5948899 DOI: 10.1186/s13287-018-0884-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/02/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
Background Peripheral nerve injury (PNI) is a worldwide issue associated with severe social and economic burden. Autologous nerve grafting, the gold standard treatment for peripheral nerve defects, still has a number of technical limitations. Tissue engineering technology is a novel therapeutic strategy, and mesenchymal stromal cells (MSCs) are promising seed cells for nerve tissue engineering. However, the efficiency of traditional methods for inducing the differentiation of MSCs to Schwann cell-like cells (SCLCs) remains unsatisfactory. Methods Here, we propose an intermittent induction method with alternate use of complete and incomplete induction medium to induce differentiation of adipose-derived stem cells (ASCs) to SCLCs. The time dependence of traditional induction methods and the efficiency of the intermittent induction method and traditional induction methods were evaluated and compared using immunocytochemistry, quantitative reverse transcription polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and co-culture with the dorsal root ganglion (DRG) in vitro. Cell transplantation was used to compare the effects of the traditional induction method and the intermittent induction method in repairing sciatic nerve defects in vivo. Results The results of the present study indicated that the intermittent induction method is more efficient than traditional methods for inducing ASCs to differentiate into SCLCs. In addition, SCLCs induced by this method were closer to mature myelinating Schwann cells and were capable of secreting neurotrophins and promoting DRG axon regeneration in vitro. Furthermore, SCLCs induced by the intermittent induction method could repair sciatic nerve defects in rats by cell transplantation in vivo more effectively than those produced by traditional methods. Conclusion Intermittent induction represents a novel strategy for obtaining seed cells for use in nerve tissue engineering. Electronic supplementary material The online version of this article (10.1186/s13287-018-0884-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xun Sun
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China.,School of Medicine, Nankai University, No.94 Weijin Road, Tianjin, 300071, People's Republic of China
| | - Yun Zhu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, No.21 Sassoon Road, Pokfulam, 999077, Hong Kong
| | - He-Yong Yin
- Department of Surgery, Experimental Surgery and Regenerative Medicine, Ludwig-Maximilians-University (LMU), Nussbaumstr. 20, 80336, Munich, Germany
| | - Zhi-Yuan Guo
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Feng Xu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Bo Xiao
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Wen-Li Jiang
- Department of Ultrasound, Beijing Hospital, National Center of Gerontology, No.1 Dahua Road, Beijing, 100730, People's Republic of China
| | - Wei-Min Guo
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Hao-Ye Meng
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Shi-Bi Lu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, People's Republic of China.
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, People's Republic of China.
| |
Collapse
|
43
|
George S, Hamblin MR, Abrahamse H. Current and Future Trends in Adipose Stem Cell Differentiation into Neuroglia. Photomed Laser Surg 2018; 36:230-240. [PMID: 29570423 DOI: 10.1089/pho.2017.4411] [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] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Neurological diseases and disorders pose a challenge for treatment and rehabilitation due to the limited capacity of the nervous system to repair itself. Adipose stem cells (ASCs) are more pliable than any adult stem cells and are capable of differentiating into non-mesodermal tissues, including neurons. Transdifferentiating ASCs to specific neuronal lineage cells enables us to deliver the right type of cells required for a replacement therapy into the nervous system. METHODS Several methodologies are being explored and tested to differentiate ASCs to functional neurons and glia with cellular factors and chemical compounds. However, none of these processes and prototypes has been wholly successful in changing the cellular structure and functional status of ASCs to become identical to neuroglial cells. In addition, successful integration and functional competence of these cells for use in clinical applications remain problematic. Photobiomodulation or low-level laser irradiation has been successfully applied to not only improve ASC viability and proliferation but has also shown promise as a possible enhancer of ASC differentiation. CONCLUSIONS Studies have shown that photobiomodulation improves the use of stem cell transplantation for neurological applications. This review investigates current neuro-differentiation inducers and suitable methodologies, including photobiomodulation, utilizing ASCs for induction of differentiation into neuronal lineages.
Collapse
Affiliation(s)
- Sajan George
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg , Doornfontein, South Africa
| | - Michael R Hamblin
- 2 Wellman Centre for Photomedicine, Massachusetts General Hospital , Boston, Massachusetts.,3 Department of Dermatology, Harvard Medical School , Boston, Massachusetts.,4 Harvard-MIT Division of Health Sciences and Technology , Cambridge, Massachusetts
| | - Heidi Abrahamse
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg , Doornfontein, South Africa
| |
Collapse
|
44
|
Tsui YP, Shea GK, Chan YS, Shum DKY. Derivation of Fate-Committed Schwann Cells from Bone Marrow Stromal Cells of Adult Rats. Methods Mol Biol 2018; 1739:137-148. [PMID: 29546705 DOI: 10.1007/978-1-4939-7649-2_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Our goal is to derive phenotypically stable Schwann cells from bone marrow stromal cells (BMSCs) for use in transplantation studies of central/peripheral nerve injuries. With the adult rat as model, here we describe steps that foster (1) expansion of the BMSC subpopulation of neural progenitors as neurosphere cells, (2) differentiation of the progenitors into Schwann cell-like cells in adherent culture supplemented with soluble factors, and (3) cell-intrinsic switch of Schwann cell-like cells to the Schwann cell fate following co-culture with sensory neurons purified from dorsal root ganglia. The derived Schwann cells retain marker expression despite withdrawal of supplements and neuronal cues, survive passaging and cryopreservation, and, importantly, show functional capacity for myelination.
Collapse
Affiliation(s)
- Y P Tsui
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Graham K Shea
- Department of Orthopaedics and Traumatology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Y S Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Daisy K Y Shum
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
45
|
Current Perspectives Regarding Stem Cell-Based Therapy for Liver Cirrhosis. Can J Gastroenterol Hepatol 2018; 2018:4197857. [PMID: 29670867 PMCID: PMC5833156 DOI: 10.1155/2018/4197857] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/16/2018] [Indexed: 12/12/2022] Open
Abstract
Liver cirrhosis is a major cause of mortality and a common end of various progressive liver diseases. Since the effective treatment is currently limited to liver transplantation, stem cell-based therapy as an alternative has attracted interest due to promising results from preclinical and clinical studies. However, there is still much to be understood regarding the precise mechanisms of action. A number of stem cells from different origins have been employed for hepatic regeneration with different degrees of success. The present review presents a synopsis of stem cell research for the treatment of patients with liver cirrhosis according to the stem cell type. Clinical trials to date are summarized briefly. Finally, issues to be resolved and future perspectives are discussed with regard to clinical applications.
Collapse
|
46
|
De la Rosa MB, Kozik EM, Sakaguchi DS. Adult Stem Cell-Based Strategies for Peripheral Nerve Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1119:41-71. [PMID: 30151648 DOI: 10.1007/5584_2018_254] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peripheral nerve injuries (PNI) occur as the result of sudden trauma and can lead to life-long disability, reduced quality of life, and heavy economic and social burdens. Although the peripheral nervous system (PNS) has the intrinsic capacity to regenerate and regrow axons to a certain extent, current treatments frequently show incomplete recovery with poor functional outcomes, particularly for large PNI. Many surgical procedures are available to halt the propagation of nerve damage, and the choice of a procedure depends on the extent of the injury. In particular, recovery from large PNI gaps is difficult to achieve without any therapeutic intervention or some form of tissue/cell-based therapy. Autologous nerve grafting, considered the "gold standard" is often implemented for treatment of gap formation type PNI. Although these surgical procedures provide many benefits, there are still considerable limitations associated with such procedures as donor site morbidity, neuroma formation, fascicle mismatch, and scarring. To overcome such restrictions, researchers have explored various avenues to improve post-surgical outcomes. The most commonly studied methods include: cell transplantation, growth factor delivery to stimulate regenerating axons and implanting nerve guidance conduits containing replacement cells at the site of injury. Replacement cells which offer maximum benefits for the treatment of PNI, are Schwann cells (SCs), which are the peripheral glial cells and in part responsible for clearing out debris from the site of injury. Additionally, they release growth factors to stimulate myelination and axonal regeneration. Both primary SCs and genetically modified SCs enhance nerve regeneration in animal models; however, there is no good source for extracting SCs and the only method to obtain SCs is by sacrificing a healthy nerve. To overcome such challenges, various cell types have been investigated and reported to enhance nerve regeneration.In this review, we have focused on cell-based strategies aimed to enhance peripheral nerve regeneration, in particular the use of mesenchymal stem cells (MSCs). Mesenchymal stem cells are preferred due to benefits such as autologous transplantation, routine isolation procedures, and paracrine and immunomodulatory properties. Mesenchymal stem cells have been transplanted at the site of injury either directly in their native form (undifferentiated) or in a SC-like form (transdifferentiated) and have been shown to significantly enhance nerve regeneration. In addition to transdifferentiated MSCs, some studies have also transplanted ex-vivo genetically modified MSCs that hypersecrete growth factors to improve neuroregeneration.
Collapse
Affiliation(s)
- Metzere Bierlein De la Rosa
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.,Veterinary Specialty Center, Buffalo Grove, IL, USA
| | - Emily M Kozik
- Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA.,Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Donald S Sakaguchi
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA. .,Biology Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA. .,Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA. .,Neuroscience Program, Iowa State University, Ames, IA, USA.
| |
Collapse
|
47
|
Moreira A, Kahlenberg S, Hornsby P. Therapeutic potential of mesenchymal stem cells for diabetes. J Mol Endocrinol 2017; 59:R109-R120. [PMID: 28739632 PMCID: PMC5570611 DOI: 10.1530/jme-17-0117] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are self-renewing multipotent cells that have the capacity to secrete multiple biologic factors that can restore and repair injured tissues. Preclinical and clinical evidence have substantiated the therapeutic benefit of MSCs in various medical conditions. Currently, MSCs are the most commonly used cell-based therapy in clinical trials because of their regenerative effects, ease of isolation and low immunogenicity. Experimental and clinical studies have provided promising results using MSCs to treat diabetes. This review will summarize the role of MSCs on tissue repair, provide emerging strategies to improve MSC function and describe how these processes translate to clinical treatments for diabetes.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of PediatricsUniversity of Texas Health Science Center-San Antonio, San Antonio, Texas, USA
| | - Samuel Kahlenberg
- Department of PediatricsUniversity of Texas Health Science Center-San Antonio, San Antonio, Texas, USA
| | - Peter Hornsby
- Department of PhysiologyTexas Research Park Campus, Barshop Institute for Longevity and Aging Studies, San Antonio, Texas, USA
| |
Collapse
|
48
|
Guo J, Guo S, Wang Y, Yu Y. Promoting potential of adipose derived stem cells on peripheral nerve regeneration. Mol Med Rep 2017; 16:7297-7304. [PMID: 28944869 PMCID: PMC5865858 DOI: 10.3892/mmr.2017.7570] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/25/2017] [Indexed: 01/08/2023] Open
Abstract
The ultimate goal of treating peripheral nerve defects is reconstructing continuity of the nerve stumps to regain nerve conduction and functional recovery. Clinically, autologous nerve grafts and Schwann cell (SC) therapy have limitations, such as the need for secondary surgery, sacrifice of another nerve and donor site complication. Adipose derived stem cells (ADSCs) may promise to be ideal alternative cells of SCs. To explore the potential of ADSCs promoting peripheral nerve regeneration, the present study investigated the influences of ADSCs on proliferation and neurotrophic function of SCs using co-culture model in vitro. Western blot analysis, immunocytochemistry, a cell viability assay, reverse transcription-polymerase chain reaction (RT-PCR) and ELISA were applied for examining the interaction of ADSCs and SCs in a co-culture model in vitro. Western blot analysis and immunocytochemistry demonstrated that protein expression levels of glial filament acidic protein (GFAP) and S100 in ADSCs co-cultured with SCs for 14 days were significantly higher compared with cells cultured alone. Cell viability assay indicated that the cell viability of SCs co-cultured with ADSCs for 3, 4, 5, 6 and 7 days was significantly higher than those cultured alone. RT-PCR showed that expression levels of neurotrophic factors [nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF)] and extracellular matrix components [fibronectin (FN) and laminin (LN)] in SCs co-cultured with ADSCs for 14 days were significantly higher than those in SCs cultured alone. NGF, GDNF, FN and LN in the supernatants of co-culture system were significantly higher than cells cultured alone, as ELISA revealed. The results of this study suggested that the transplantation of ADSCs may have a promoting potential to the peripheral nerve regeneration as undifferentiated state.
Collapse
Affiliation(s)
- Jiayan Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shu Guo
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuxin Wang
- Department of Plastic Surgery, First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yanqiu Yu
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
49
|
Directed Differentiation of Human Bone Marrow Stromal Cells to Fate-Committed Schwann Cells. Stem Cell Reports 2017; 9:1097-1108. [PMID: 28890164 PMCID: PMC5639182 DOI: 10.1016/j.stemcr.2017.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022] Open
Abstract
Our ultimate goal of in vitro derivation of Schwann cells (SCs) from adult bone marrow stromal cells (BMSCs) is such that they may be used autologously to assist post-traumatic nerve regeneration. Existing protocols for derivation of SC-like cells from BMSCs fall short in the stability of the acquired phenotype and the functional capacity to myelinate axons. Our experiments indicated that neuro-ectodermal progenitor cells among the human hBMSCs could be selectively expanded and then induced to differentiate into SC-like cells. Co-culture of the SC-like cells with embryonic dorsal root ganglion neurons facilitated contact-mediated signaling that accomplished the switch to fate-committed SCs. Microarray analysis and in vitro myelination provided evidence that the human BMSC-derived SCs were functionally mature. This was reinforced by repair and myelination phenotypes observable in vivo with the derived SCs seeded into a nerve guide as an implant across a critical gap in a rat model of sciatic nerve injury. A protocol for in vitro derivation of fate-committed SCs from human BMSCs The derived human SCs were functionally capable of myelination in vitro The derived human SCs guided axonal regrowth and formed compact myelin in vivo
Collapse
|
50
|
|