1
|
Khaled MM, Ibrahium AM, Abdelgalil AI, El-Saied MA, El-Bably SH. Regenerative Strategies in Treatment of Peripheral Nerve Injuries in Different Animal Models. Tissue Eng Regen Med 2023; 20:839-877. [PMID: 37572269 PMCID: PMC10519924 DOI: 10.1007/s13770-023-00559-4] [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: 03/28/2023] [Revised: 05/15/2023] [Accepted: 05/21/2023] [Indexed: 08/14/2023] Open
Abstract
BACKGROUND Peripheral nerve damage mainly resulted from traumatic or infectious causes; the main signs of a damaged nerve are the loss of sensory and/or motor functions. The injured nerve has limited regenerative capacity and is recovered by the body itself, the recovery process depends on the severity of damage to the nerve, nowadays the use of stem cells is one of the new and advanced methods for treatment of these problems. METHOD Following our review, data are collected from different databases "Google scholar, Springer, Elsevier, Egyptian Knowledge Bank, and PubMed" using different keywords such as Peripheral nerve damage, Radial Nerve, Sciatic Nerve, Animals, Nerve regeneration, and Stem cell to investigate the different methods taken in consideration for regeneration of PNI. RESULT This review contains tables illustrating all forms and types of regenerative medicine used in treatment of peripheral nerve injuries (PNI) including different types of stem cells " adipose-derived stem cells, bone marrow stem cells, Human umbilical cord stem cells, embryonic stem cells" and their effect on re-constitution and functional recovery of the damaged nerve which evaluated by physical, histological, Immuno-histochemical, biochemical evaluation, and the review illuminated the best regenerative strategies help in rapid peripheral nerve regeneration in different animal models included horse, dog, cat, sheep, monkey, pig, mice and rat. CONCLUSION Old surgical attempts such as neurorrhaphy, autogenic nerve transplantation, and Schwann cell implantation have a limited power of recovery in cases of large nerve defects. Stem cell therapy including mesenchymal stromal cells has a high potential differentiation capacity to renew and form a new nerve and also restore its function.
Collapse
Affiliation(s)
- Mona M Khaled
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt.
| | - Asmaa M Ibrahium
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Ahmed I Abdelgalil
- Department of Surgery, Anaesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Mohamed A El-Saied
- Department of Pathology, Faculty of Veterinary of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| | - Samah H El-Bably
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza Square, Giza, 12211, Egypt
| |
Collapse
|
2
|
Natarajan A, Sethumadhavan A, Krishnan UM. Toward Building the Neuromuscular Junction: In Vitro Models To Study Synaptogenesis and Neurodegeneration. ACS OMEGA 2019; 4:12969-12977. [PMID: 31460423 PMCID: PMC6682064 DOI: 10.1021/acsomega.9b00973] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
The neuromuscular junction (NMJ) is a unique, specialized chemical synapse that plays a crucial role in transmitting and amplifying information from spinal motor neurons to skeletal muscles. NMJ complexity ensures closely intertwined interactions between numerous synaptic vesicles, signaling molecules, ion channels, motor neurons, glia, and muscle fibers, making it difficult to dissect the underlying mechanisms and factors affecting neurodegeneration and muscle loss. Muscle fiber or motor neuron cell death followed by rapid axonal degeneration due to injury or disease has a debilitating effect on movement and behavior, which adversely affects the quality of life. It thus becomes imperative to study the synapse and intercellular signaling processes that regulate plasticity at the NMJ and elucidate mechanisms and pathways at the cellular level. Studies using in vitro 2D cell cultures have allowed us to gain a fundamental understanding of how the NMJ functions. However, they do not provide information on the intricate signaling networks that exist between NMJs and the biological environment. The advent of 3D cell cultures and microfluidic lab-on-a-chip technologies has opened whole new avenues to explore the NMJ. In this perspective, we look at the challenges involved in building a functional NMJ and the progress made in generating models for studying the NMJ, highlighting the current and future applications of these models.
Collapse
Affiliation(s)
- Anupama Natarajan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Anjali Sethumadhavan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Uma Maheswari Krishnan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| |
Collapse
|
3
|
Ruven C, Badea SR, Wong WM, Wu W. Combination Treatment With Exogenous GDNF and Fetal Spinal Cord Cells Results in Better Motoneuron Survival and Functional Recovery After Avulsion Injury With Delayed Root Reimplantation. J Neuropathol Exp Neurol 2019; 77:325-343. [PMID: 29420729 DOI: 10.1093/jnen/nly009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
When spinal roots are torn off from the spinal cord, both the peripheral and central nervous system get damaged. As the motoneurons lose their axons, they start to die rapidly, whereas target muscles atrophy due to the denervation. In this kind of complicated injury, different processes need to be targeted in the search for the best treatment strategy. In this study, we tested glial cell-derived neurotrophic factor (GDNF) treatment and fetal lumbar cell transplantation for their effectiveness to prevent motoneuron death and muscle atrophy after the spinal root avulsion and delayed reimplantation. Application of exogenous GDNF to injured spinal cord greatly prevented the motoneuron death and enhanced the regeneration and axonal sprouting, whereas no effect was seen on the functional recovery. In contrast, cell transplantation into the distal nerve did not affect the host motoneurons but instead mitigated the muscle atrophy. The combination of GDNF and cell graft reunited the positive effects resulting in better functional recovery and could therefore be considered as a promising strategy for nerve and spinal cord injuries that involve the avulsion of spinal roots.
Collapse
Affiliation(s)
- Carolin Ruven
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | - Wai-Man Wong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wutian Wu
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.,GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Re-Stem Biotechnology Co., Ltd, Jiangsu, China
| |
Collapse
|
4
|
Zheng Y, Huang C, Liu F, Lin H, Niu Y, Yang X, Zhang Z. Reactivation of denervated Schwann cells by neurons induced from bone marrow-derived mesenchymal stem cells. Brain Res Bull 2018. [DOI: 10.1016/j.brainresbull.2018.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
5
|
Sharma A, Sane H, Gokulchandran N, Badhe P, Pai S, Kulkarni P, Yadav J, Inamdar S. Cellular Therapy for Chronic Traumatic Brachial Plexus Injury. Adv Biomed Res 2018; 7:51. [PMID: 29657936 PMCID: PMC5887704 DOI: 10.4103/2277-9175.228631] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Cellular therapy is being actively pursued as a therapeutic modality in many of the neurological diseases. A variety of stem cells from diverse sources have been studied in detail and have been shown to exhibit angiogenetic and immunomodulatory properties in addition to other neuroprotective effects. Published clinical data have shown bone marrow mononuclear cell (BMMNC) injection in neurological disorders is safe and possesses regenerative potential. We illustrate a case of 27-year-old male with traumatic brachial plexus injury, administered with autologous BMMNCs intrathecally and intramuscularly, followed by multidisciplinary rehabilitation. At the follow-up assessment of 3 and 7 months after first cell transplantation, improvements were recorded in muscle strength and movements. Electromyography (EMG) performed after the intervention showed a response in biceps and deltoid muscles suggesting the process of reinnervation at the site of injury. In view of the improvements observed after the treatment, the patient underwent second cell transplantation 8 months after the first transplantation. Muscle wasting had completely stopped with an increase in the muscle girth. No adverse effects were noted. Improvements were maintained for 4 years. A comprehensive randomized study for this type of injury is needed to establish the therapeutic benefits of cellular therapy.
Collapse
Affiliation(s)
- Alok Sharma
- Department of Medical Services and Clinical Research, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Hemangi Sane
- Department of Research and Development, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Nandini Gokulchandran
- Department of Medical Services and Clinical Research, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Prerna Badhe
- Department of Medical Services and Clinical Research, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Suhasini Pai
- Department of Research and Development, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Pooja Kulkarni
- Department of Research and Development, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Jayanti Yadav
- Department of Neurorehabilitation, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| | - Sanket Inamdar
- Department of Neurorehabilitation, NeuroGen Brain and Spine Institute, New Mumbai, Maharashtra, India
| |
Collapse
|
6
|
Yu Z, Men Y, Dong P. Schwann cells promote the capability of neural stem cells to differentiate into neurons and secret neurotrophic factors. Exp Ther Med 2017; 13:2029-2035. [PMID: 28565804 PMCID: PMC5443174 DOI: 10.3892/etm.2017.4183] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/20/2017] [Indexed: 11/05/2022] Open
Abstract
The present study investigated whether co-culturing Schwann cells (SCs) with neural stem cells (NSCs) improves viability, direction of differentiation and secretion of brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor (GDNF) in NSCs. The three groups assessed were as follows: SCs, NSCs, and a co-culture of SCs and NSCs. Cellular morphological changes were observed under an inverted phase contrast microscope and quantified. Cells were identified by immunofluorescence staining: S100 for SCs, Nestin for NSCs, microtubule associated protein (Map) 2 and NeuN for neurons and glial fibrillary acidic protein for astrocytes. Cell viability was evaluated by MTT assay. Secretion of BDNF and GDNF was quantified; mRNA expression was quantified by reverse transcription-quantitative polymerase chain reaction. The majority of NSCs in the co-cultured group differentiated into neurons. The cell survival rate of the co-culture group was significantly higher than the other groups on days 3, 5 and 10 (P<0.01). The secretion of BDNF in the co-culture group was significantly higher than NSCs on days 3, 5 and 7 (P<0.05), while the amount of GDNF in co-culture was significantly higher than both NSCs and SCs on day 1 (P<0.05). BDNF and GDNF gene expression in the co-culture group was significantly higher than SCs (P<0.01). Gene expression of Map2 in co-culture group was also significantly higher than both NSC and SC groups (P<0.01). Therefore, co-cultured SCs and NSCs promote differentiation of NSCs into neurons and secrete higher levels of neurotropic factors including BDNF and GDNF.
Collapse
Affiliation(s)
- Ziwei Yu
- Department of Otolaryngology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Yongzhi Men
- Department of Otolaryngology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| | - Pin Dong
- Department of Otolaryngology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P.R. China
| |
Collapse
|
7
|
Ruven C, Li W, Li H, Wong WM, Wu W. Transplantation of Embryonic Spinal Cord Derived Cells Helps to Prevent Muscle Atrophy after Peripheral Nerve Injury. Int J Mol Sci 2017; 18:ijms18030511. [PMID: 28264437 PMCID: PMC5372527 DOI: 10.3390/ijms18030511] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Injuries to peripheral nerves are frequent in serious traumas and spinal cord injuries. In addition to surgical approaches, other interventions, such as cell transplantation, should be considered to keep the muscles in good condition until the axons regenerate. In this study, E14.5 rat embryonic spinal cord fetal cells and cultured neural progenitor cells from different spinal cord segments were injected into transected musculocutaneous nerve of 200–300 g female Sprague Dawley (SD) rats, and atrophy in biceps brachii was assessed. Both kinds of cells were able to survive, extend their axons towards the muscle and form neuromuscular junctions that were functional in electromyographic studies. As a result, muscle endplates were preserved and atrophy was reduced. Furthermore, we observed that the fetal cells had a better effect in reducing the muscle atrophy compared to the pure neural progenitor cells, whereas lumbar cells were more beneficial compared to thoracic and cervical cells. In addition, fetal lumbar cells were used to supplement six weeks delayed surgical repair after the nerve transection. Cell transplantation helped to preserve the muscle endplates, which in turn lead to earlier functional recovery seen in behavioral test and electromyography. In conclusion, we were able to show that embryonic spinal cord derived cells, especially the lumbar fetal cells, are beneficial in the treatment of peripheral nerve injuries due to their ability to prevent the muscle atrophy.
Collapse
Affiliation(s)
- Carolin Ruven
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.
| | - Wen Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.
| | - Heng Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.
| | - Wai-Man Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.
| | - Wutian Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Joint Laboratory for CNS Regeneration, Jinan University and The University of Hong Kong, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510000, China.
- Guangdong Engineering Research Center of Stem Cell Storage and Clinical Application, Saliai Stem Cell Science and Technology, Guangzhou 510000, China.
| |
Collapse
|
8
|
Wang C, Lu CF, Peng J, Hu CD, Wang Y. Roles of neural stem cells in the repair of peripheral nerve injury. Neural Regen Res 2017; 12:2106-2112. [PMID: 29323053 PMCID: PMC5784362 DOI: 10.4103/1673-5374.221171] [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] [Indexed: 01/09/2023] Open
Abstract
Currently, researchers are using neural stem cell transplantation to promote regeneration after peripheral nerve injury, as neural stem cells play an important role in peripheral nerve injury repair. This article reviews recent research progress of the role of neural stem cells in the repair of peripheral nerve injury. Neural stem cells can not only differentiate into neurons, astrocytes and oligodendrocytes, but can also differentiate into Schwann-like cells, which promote neurite outgrowth around the injury. Transplanted neural stem cells can differentiate into motor neurons that innervate muscles and promote the recovery of neurological function. To promote the repair of peripheral nerve injury, neural stem cells secrete various neurotrophic factors, including brain-derived neurotrophic factor, fibroblast growth factor, nerve growth factor, insulin-like growth factor and hepatocyte growth factor. In addition, neural stem cells also promote regeneration of the axonal myelin sheath, angiogenesis, and immune regulation. It can be concluded that neural stem cells promote the repair of peripheral nerve injury through a variety of ways.
Collapse
Affiliation(s)
- Chong Wang
- Central Hospital of Handan, Handan, Hebei Province; Institute of Orthopedics, Chinese PLA General Hospital, Beijing; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Chang-Feng Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, ; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries of Chinese PLA, Beijing, China
| | - Cheng-Dong Hu
- Central Hospital of Handan, Handan, Hebei Province, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries of Chinese PLA, Beijing, China
| |
Collapse
|
9
|
Javanmardy S, Asadi MH, Movahedin M, Moradpour F, Bahadoran H. Derivation of motor neuron-like cells from neonatal mouse testis in a simple culture condition. Andrologia 2016; 48:1100-1107. [PMID: 26892722 DOI: 10.1111/and.12545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2015] [Indexed: 11/28/2022] Open
Abstract
Embryonic stem cell (ESC) therapy is an exciting way to treat neurodegenerative disease and central nervous system injury. However, many ethical and immunological problems surround the use of embryonic stem cells. Finding an alternative source of stem cells is therefore pertinent. In this study, spermatogonia stem cells (SSCs) were used to generate mature motor neurons. SSCs were extracted from neonatal testes and cultured in DMED/F12 medium for 3 weeks. Characterisation of SSC-derived ESC-like cells was confirmed by RT-qPCR, immunostaining, alkaline phosphatase activity and their ability to form embryoid bodies (EBs). The EBs were induced by retinoic acid and Sonic hedgehog and trypsinised to obtain single induced cells. The single cells were cultured in neural medium for 18 days. Characterisation of neural precursors and motor neuron-like cells was confirmed by RT-qPCR and immunocytochemical analysis at the 7th day (early stage) and 18th day (late stage), respectively, of culturing. The neural precursors were found to be positive for nestin and Sox2, and a small fraction of cells expressed β-tubulin III. Upon further differentiation, multipolar neurons were detected that expressed β-tubulin III and MAP2 markers. Moreover, the expression levels of Olig2 and PAX6 were significantly lower, while HB9, Isl1 and Isl2 expression levels were higher at the late stage when compared to the early stage. These results show that SSCs have the potential to differentiate to motor neuron-like cells and express markers specific for mature motor neurons. However, the functional ability of these cells remains to be evaluated in future studies.
Collapse
Affiliation(s)
- S Javanmardy
- Department of Midwifery, School of Nursing & Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - M H Asadi
- Department of Anatomical Sciences, Faculty of medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - M Movahedin
- Department of Anatomical Sciences, Faculty of medical sciences, Tarbiat Modares University, Tehran, Iran
| | - F Moradpour
- Department of Physiology & Pharmacology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - H Bahadoran
- Department of Anatomical Sciences, Faculty of medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Liu F, Lin H, Zhang C. Construction of Tissue-Engineered Nerve Conduits Seeded with Neurons Derived from Hair-Follicle Neural Crest Stem Cells. Methods Mol Biol 2016; 1453:33-38. [PMID: 27431244 DOI: 10.1007/978-1-4939-3786-8_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Tissue-engineered nerve conduits are widely used for the study of peripheral nerve injury repair. With regard to repairing long nerve defects, stem-cell-derived neurons are recommended as seed cells. As hair-follicle neural crest stem cells (hfNCSCs) are easily to be harvested from patients and have the potential to differentiate into neuronal cells, hfNCSCs-derived neurons are an ideal candidate choice. Acellular nerve grafts, a type of biological material scaffold, with intact collagen structure, with biocompatibility and less toxicity are obtained through removing live cells with 1 % lysolecithin, are also an ideal choice. In the present report, we describe a tissue-engineered nerve conduit seeded with rat hfNCSCs-derived neurons into the beagle acellular sciatic nerve scaffold. Our goal is to provide a novel engineered therapeutic for repairing peripheral nerve injury with long distance defects.
Collapse
Affiliation(s)
- Fang Liu
- Department of Anatomy, Institute of Biomedical Engineering, Second Military Medical University, Shanghai, People's Republic of China.
- AntiCancer, Inc., San Diego, CA, USA.
| | - Haiyan Lin
- Department of Anatomy, Institute of Biomedical Engineering, Second Military Medical University, Shanghai, People's Republic of China
| | - Chuansen Zhang
- Department of Anatomy, Institute of Biomedical Engineering, Second Military Medical University, Shanghai, People's Republic of China.
| |
Collapse
|
11
|
Bone marrow-derived, neural-like cells have the characteristics of neurons to protect the peripheral nerve in microenvironment. Stem Cells Int 2015; 2015:941625. [PMID: 25861281 PMCID: PMC4378708 DOI: 10.1155/2015/941625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/27/2015] [Indexed: 11/18/2022] Open
Abstract
Effective repair of peripheral nerve defects is difficult because of the slow growth of new axonal growth. We propose that "neural-like cells" may be useful for the protection of peripheral nerve destructions. Such cells should prolong the time for the disintegration of spinal nerves, reduce lesions, and improve recovery. But the mechanism of neural-like cells in the peripheral nerve is still unclear. In this study, bone marrow-derived neural-like cells were used as seed cells. The cells were injected into the distal end of severed rabbit peripheral nerves that were no longer integrated with the central nervous system. Electromyography (EMG), immunohistochemistry, and transmission electron microscopy (TEM) were employed to analyze the development of the cells in the peripheral nerve environment. The CMAP amplitude appeared during the 5th week following surgery, at which time morphological characteristics of myelinated nerve fiber formation were observed. Bone marrow-derived neural-like cells could protect the disintegration and destruction of the injured peripheral nerve.
Collapse
|
12
|
El Maarouf A, Yaw DML, Rutishauser U. Improved stem cell-derived motoneuron survival, migration, sprouting, and innervation with enhanced expression of polysialic acid. Cell Transplant 2014; 24:797-809. [PMID: 24593882 DOI: 10.3727/096368914x679228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Motoneurons (MNs) derived from mouse embryonic stem cells (ESCs) begin to express low levels of polysialic acid (PSA) at the time when they acquire an ability to migrate and extend neurites. PSA is known to promote cell migration and process outgrowth/guidance in the developing nervous system. To test if experimentally enhanced expression of PSA would augment these cellular events, the PSA-synthesizing polysialyltransferase was introduced into ESCs. In culture, the resulting higher PSA expression specifically increased neurite outgrowth and cell migration from differentiated embryoid bodies. In addition, the MN population obtained after sorting for HB9::GFP expression showed enhanced survival as well as extensive neurite outgrowth. Following transplantation of ESC-derived MNs into an adult sciatic nerve devoid of endogenous axons, the PSA augmentation increased the numbers of axons growing toward the denervated muscles. Migration of some transplanted cells inside the nerve toward muscle was also enhanced. Moreover, higher PSA expression selectively affected target innervation. It produced greater numbers of neuromuscular junctions in a predominantly fast twitch muscle and had no effect in a slow twitch muscle. These findings suggest that engineering of PSA expression in ESC could serve as an enhancement for MN cell therapy.
Collapse
Affiliation(s)
- Abderrahman El Maarouf
- Cellular and Developmental Neuroscience, Department of Cell Biology, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
13
|
Comparison of different protocols for neural differentiation of human induced pluripotent stem cells. Mol Biol Rep 2014; 41:1713-21. [DOI: 10.1007/s11033-014-3020-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/02/2014] [Indexed: 12/27/2022]
|
14
|
Su H, Zhang W, Yang X, Qin D, Sang Y, Wu C, Wong WM, Yuan Q, So KF, Wu W. Neural Progenitor Cells Generate Motoneuron-Like Cells to Form Functional Connections with Target Muscles after Transplantation into the Musculocutaneous Nerve. Cell Transplant 2012; 21:2651-63. [DOI: 10.3727/096368912x654975] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neural progenitor cells (NPCs) are suggested to be a valuable source of cell transplant in treatment of various neurological diseases because of their distinct attributes. They can be expanded and induced to differentiate in vitro. However, it remains uncertain whether in vitro expanded NPCs have the capacity to give rise to functional motoneurons after transplantation in vivo. Here, we showed that in vitro expanded NPCs, when transplanted into the musculocutaneous nerve, generated motoneuron-like cells that exhibited typical morphology with large cell bodies, expressed specific molecules, and extended axons to form functional connections with the target muscle. In contrast, transplanted NPCs failed to yield motoneurons in the injured ventral horn of the spinal cord. The results of the study demonstrate that NPCs have the potential to generate functional motoneurons in an appropriate environment. The distinct differentiating fate of NPCs in the musculocutaneous nerve and the injured ventral horn suggests the importance and necessity of modifying the host microenvironment in use of NPCs for cell replacement therapies for motoneuron diseases.
Collapse
Affiliation(s)
- Huanxing Su
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wenming Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Xiaoying Yang
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Dajiang Qin
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yanhua Sang
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chaoyang Wu
- Department of Orthopaedics, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Wai-Man Wong
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Qiuju Yuan
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kwok-Fai So
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Joint Laboratory for Brain Function and Health (BFAH), Jinan University and The University of Hong Kong, Jinan University, Guangzhou, China
| | - Wutian Wu
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Joint Laboratory for Brain Function and Health (BFAH), Jinan University and The University of Hong Kong, Jinan University, Guangzhou, China
- Research Center of Reproduction, Development and Growth, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
15
|
Lin S, Xu L, Hu S, Zhang C, Wang Y, Xu J. Optimal time-point for neural stem cell transplantation to delay denervated skeletal muscle atrophy. Muscle Nerve 2012; 47:194-201. [PMID: 23042154 DOI: 10.1002/mus.23447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2012] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Transplantation of neural stem cells (NSCs) is a promising treatment to delay denervated skeletal muscle atrophy; however, the optimal time-point between peripheral nerve injury and NSC transplantation needs to be determined. METHODS Improvement in rat gastrocnemius muscle function was evaluated after NSCs were transplanted into sectioned distal tibial nerves. We also assessed survival and differentiation. ANOVA was used to compare the mean value of the number of neuron-like cells, cross-sectional area amelioration, the amount of activated fibers, and latency and amplitude of the gastrocnemius compound muscle action potential. RESULTS The group in which the NSCs were transplanted 1 week after tibial nerve transection had the largest number of neuron-like cells, maximum cross-sectional area amelioration, and maximum amount of activated gastrocnemius fibers compared with all other groups (P < 0.01). CONCLUSIONS The optimal time-point for NSC transplantation for delaying denervated skeletal muscle atrophy is 1 week after severing the nerve.
Collapse
Affiliation(s)
- Sen Lin
- Department of Orthopaedics, Shanghai Sixth People's Hospital and School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
16
|
Dadon-Nachum M, Melamed E, Offen D. Stem cells treatment for sciatic nerve injury. Expert Opin Biol Ther 2011; 11:1591-7. [DOI: 10.1517/14712598.2011.628933] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
17
|
Luu B, Ellisor D, Zervas M. The lineage contribution and role of Gbx2 in spinal cord development. PLoS One 2011; 6:e20940. [PMID: 21698205 PMCID: PMC3116860 DOI: 10.1371/journal.pone.0020940] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 05/16/2011] [Indexed: 12/30/2022] Open
Abstract
Background Forging a relationship between progenitors with dynamically changing gene expression and their terminal fate is instructive for understanding the logic of how cell-type diversity is established. The mouse spinal cord is an ideal system to study these mechanisms in the context of developmental genetics and nervous system development. Here we focus on the Gastrulation homeobox 2 (Gbx2) transcription factor, which has not been explored in spinal cord development. Methodology/Principal Findings We determined the molecular identity of Gbx2-expressing spinal cord progenitors. We also utilized genetic inducible fate mapping to mark the Gbx2 lineage at different embryonic stages in vivo in mouse. Collectively, we uncover cell behaviors, cytoarchitectonic organization, and the terminal cell fate of the Gbx2 lineage. Notably, both ventral motor neurons and interneurons are derived from the Gbx2 lineage, but only during a short developmental period. Short-term fate mapping during mouse spinal cord development shows that Gbx2 expression is transient and is extinguished ventrally in a rostral to caudal gradient. Concomitantly, a permanent lineage restriction boundary ensures that spinal cord neurons derived from the Gbx2 lineage are confined to a dorsal compartment that is maintained in the adult and that this lineage generates inhibitory interneurons of the spinal cord. Using lineage tracing and molecular markers to follow Gbx2-mutant cells, we show that the loss of Gbx2 globally affects spinal cord patterning including the organization of interneuron progenitors. Finally, long-term lineage analysis reveals that the presence and timing of Gbx2 expression in interneuron progenitors results in the differential contribution to subtypes of terminally differentiated interneurons in the adult spinal cord. Conclusions/Significance We illustrate the complex cellular nature of Gbx2 expression and lineage contribution to the mouse spinal cord. In a broader context, this study provides a direct link between spinal cord progenitors undergoing dynamic changes in molecular identity and terminal neuronal fate.
Collapse
Affiliation(s)
- Brian Luu
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Debra Ellisor
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Mark Zervas
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
18
|
Cheng LN, Duan XH, Zhong XM, Guo RM, Zhang F, Zhou CP, Shen J. Transplanted neural stem cells promote nerve regeneration in acute peripheral nerve traction injury: assessment using MRI. AJR Am J Roentgenol 2011; 196:1381-1387. [PMID: 21606303 DOI: 10.2214/ajr.10.5495] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The purpose of our study was to monitor neural stem cells (NSCs) transplanted in acute peripheral nerve traction injury and to use MRI to assess the ability of NSCs to promote nerve regeneration. MATERIALS AND METHODS After labeling with gadolinium-diethylene triamine pentaacetic acid (gadopentetate dimeglumine) and fluorescent dye (PKH26), 5 × 10(5) NSCs were grafted to acutely distracted sciatic nerves in 21 New Zealand White rabbits. In addition, 5 × 10(5) unlabeled NSCs (n = 21) and vehicle alone (n = 21) subjects were injected as a control. Serial MRI was performed with a 1.5-T scanner to determine the distribution of grafted cells. Sequential T1 and T2 values of the nerves and functional recovery were measured over a 70-day follow-up period, with histologic assessments performed at regular intervals. RESULTS The distribution and migration of labeled NSCs could be tracked with MRI until 10 days after transplantation. Compared with vehicle control, nerves grafted with labeled or unlabeled NSCs had better functional recovery and showed improved nerve regeneration but exhibited a sustained increase of T1 and T2 values during the phase of regeneration. CONCLUSION Gadopentetate dimeglumine-based labeling allowed short-term in vivo MRI tracking of NSCs grafted in injured nerves. NSCs transplantation could promote nerve regeneration in acute peripheral nerve traction injury as shown by a prolonged increase of nerve T1 and T2 values.
Collapse
Affiliation(s)
- Li-Na Cheng
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang Rd West, Guangzhou 510120, Guangdong, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Gu SH, Xu WD, Xu L, Li XK, Ochiya T, Wang Y, Li JF, Gu YD, Xu JG. Regenerated host axons form synapses with neurons derived from neural stem cells transplanted into peripheral nerves. J Int Med Res 2011; 38:1721-9. [PMID: 21309486 DOI: 10.1177/147323001003800517] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
It is reported that neural stem cells (NSC) can arrest denervated muscle atrophy and promote nerve regeneration when transplanted into injured peripheral nerves, and that regenerated host axons can form synapses with transplanted and differentiated NSC. In this study, F344 rat nerve segments and F344 rat NSC were transplanted into host green fluorescence protein (GFP) transgenic F344 rats. This allowed transplanted F344 rat tissue to be used as a nonluminous background for the clear visualization of regenerated host GFP axons. Regenerated host axons grew into the transplanted F344 nerve segment 2 weeks after nerve anastomosis. Immunohistochemical staining and confocal microscope analysis revealed that regenerated host axons formed synapses with NSC-derived neurons. The findings confirmed that regenerated peripheral axons form synapses with neurons in peripheral nerves, possibly forming the basis for clinical application in peripheral nerve injury.
Collapse
Affiliation(s)
- S H Gu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gu S, Shen Y, Xu W, Xu L, Li X, Zhou G, Gu Y, Xu J. Application of fetal neural stem cells transplantation in delaying denervated muscle atrophy in rats with peripheral nerve injury. Microsurgery 2010; 30:266-74. [PMID: 19967766 DOI: 10.1002/micr.20722] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Injury to peripheral nerves always results in progressive skeletal muscle atrophy and poor functional recovery. Previous studies have demonstrated that transplanting neural stem cells (NSCs) into peripheral nerve can differentiate into neurons and delay muscle atrophy. However, the mechanism was not very clear. In this study, we transplanted the fetal NSCs to the injured nerve and examined new formed neuromuscular junctions (NMJs) in the denervated muscle and arrest of muscle atrophy. In our study, two pregnant Fischer rats were used to harvest fetal NSCs, 70 rats were randomly divided into NSC-transplanted and control groups, five rats without surgery were used as the normal control. A volume of 5 microl culture media with or without fetal NSCs (5 x 10(6)) were transplanted into distal tibial nerve stump after the nerve was transected in two groups, respectively. Three, five, and seven months after denervation, the dry weight of gastrocnemius muscle was found significant heavier, and the fiber area was more retained in NSC-transplanted group comparing to the control group (P < 0.05). Neurons were found in the distal tibial nerves even 7 months after fetal NSCs transplantation. Newly formed NMJs were detected by immunohistochemistry. In addition, the results of electrophysiological analysis and retrograde tracing manifested that the neural pathway between muscle and differentiated neurons was integrity. In conclusions, our study demonstrated that fetal NSCs transplanted into peripheral nerves could differentiate into neurons and form functional NMJs with denervated muscle, which may be beneficial for the treatment of muscle atrophy after peripheral nerve injury.
Collapse
Affiliation(s)
- Shihui Gu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Petrova ES. Studies of histogenetic and neurodegenerative processes in the nervous system using heterotopic neurotransplantation. ACTA ACUST UNITED AC 2010; 40:823-32. [PMID: 20680478 DOI: 10.1007/s11055-010-9333-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Indexed: 12/25/2022]
Abstract
The aim of the present article is to summarize our own experimental and published data on neurotransplantation into ectopic sites such as peripheral nerves (mainly) and the anterior chamber of the eye in rats. The review addresses questions touching on the following problems: the histogenesis and survival of nervous tissue after transplantation, the interaction between transplanted tissues with recipient tissues, assessment of long-term living transplants, simultaneous transplantation of different embryonic rudiments, transplantation of spinal cord ganglia, and the effects of trophic factors on the development of transplants. New data on stem cell transplantation into peripheral nerves are discussed.
Collapse
Affiliation(s)
- E S Petrova
- Department of General and Specific Morphology, Research Institute of Experimental Medicine, North West Branch, Russian Academy of Medical Sciences, St. Petersburg, Russia
| |
Collapse
|
22
|
Abstract
OBJECTIVE The purpose of this review is to summarize the basic science literature related to chronic nerve injuries, and to then use this as the background to provide emerging insights into the promising role of cellular therapy for nerve injury repair. METHODS The literature pertinent to the experimental and clinical aspects of chronic nerve injury was reviewed, as was emerging literature and our own recent experience in using cellular therapy to repair injured nerves. RESULTS Peripheral nerves have the potential to regenerate axons and reinnervate end organs. Yet, outcome after peripheral nerve injury, even after nerve repair, remains relatively poor. The single most important quantitative contributor to poor motor recovery is chronic denervation of the distal nerve. Chronic denervation is common because of the often extensive injury zone that prevents any axonal outgrowth or (even if outgrowth occurs) the relatively slow rate of regeneration. As a consequence, the distal nerve remains chronically devoid of regrowing axons. In turn, prolonged denervation of Schwann cells (SCs) seems to be the critical factor that makes them unreceptive for axonal regeneration. Regenerative success was demonstrated when denervated SCs were replaced with healthy SCs cultured from a secondary nerve. This cell-replacement strategy is, however, limited in the clinical setting by the inability to obtain sufficient numbers of cells and the requirement for sacrifice of additional nerve tissue. We, along with several other groups, have therefore begun investigating stem cell therapies to improve the regenerative environment. CONCLUSION There are several avenues of stem cell-based approaches to peripheral nerve repair. One of these, skin-derived precursor cells, are easily accessible, autologous adult stem cells that can survive and myelinate in the peripheral nerve environment and become SC-like in their apparent differentiation.
Collapse
Affiliation(s)
- Sarah Walsh
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | | |
Collapse
|
23
|
Ymlahi-Ouazzani Q, J Bronchain O, Paillard E, Ballagny C, Chesneau A, Jadaud A, Mazabraud A, Pollet N. Reduced levels of survival motor neuron protein leads to aberrant motoneuron growth in a Xenopus model of muscular atrophy. Neurogenetics 2009; 11:27-40. [PMID: 19517146 DOI: 10.1007/s10048-009-0200-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 05/20/2009] [Indexed: 10/20/2022]
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by motor neuron loss and skeletal muscle atrophy. The loss of function of the smn1 gene, the main supplier of survival motor neuron protein (SMN) protein in human, leads to reduced levels of SMN and eventually to SMA. Here, we ask if the amphibian Xenopus tropicalis can be a good model system to study SMA. Inhibition of the production of SMN using antisense morpholinos leads to caudal muscular atrophy in tadpoles. Of note, early developmental patterning of muscles and motor neurons is unaffected in this system as well as acetylcholine receptors clustering. Muscular atrophy seems to rather result from aberrant pathfinding and growth arrest and/or shortening of motor axons. This event occurs in the absence of neuronal cell bodies apoptosis, a process comparable to that of amyotrophic lateral sclerosis. Xenopus tropicalis is revealed as a complementary animal model for the study of SMA.
Collapse
Affiliation(s)
- Qods Ymlahi-Ouazzani
- CNRS UMR 8080, Laboratoire Développement, Morphogenèse et Evolution, 91405, Orsay, France
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Transplanted mouse embryonic stem-cell-derived motoneurons form functional motor units and reduce muscle atrophy. J Neurosci 2009; 28:12409-18. [PMID: 19020033 DOI: 10.1523/jneurosci.1761-08.2008] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prolonged muscle denervation resulting from motor neuron (MN) damage leads to atrophy and degeneration of neuromuscular junctions (NMJs), which can impart irreversible damage. In this study, we ask whether transplanted embryonic stem (ES) cells differentiated into MNs can form functional synapses with host muscle, and if so what effects do they have on the muscle. After transplantation into transected tibial nerves of adult mice, ES-cell-derived MNs formed functional synapses with denervated host muscle, which resulted in the ability to produce average tetanic forces of 44% of nonlesioned controls. ES-cell-derived motor units (MUs) had mean force values and ranges similar to control muscles. The number of type I fibers and fatigue resistance of the MUs were increased, and denervation-associated muscle atrophy was significantly reduced. These results demonstrate the capacity for ES-cell-derived MNs not only to incorporate into the adult host tissue, but also to exert changes in the target tissue. By providing the signals normally active during embryonic development and placing the cells in an environment with their target tissue, ES cells differentiate into MNs that give rise to functional MU output which resembles the MU output of endogenous MNs. This suggests that these signals combined with those present in the graft environment, lead to the activation of a program intended to produce a normal range of MU forces.
Collapse
|
25
|
Xiong G, Ozaki N, Sugiura Y. Transplanted embryonic spinal tissue promotes severed sciatic nerve regeneration in rats. ACTA ACUST UNITED AC 2009; 72:127-38. [DOI: 10.1679/aohc.72.127] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ge Xiong
- Department of Hand Surgery, Beijing Jishuitan Hospital
| | - Noriyuki Ozaki
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine
| | - Yasuo Sugiura
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine
| |
Collapse
|
26
|
Grosheva M, Guntinas-Lichius O, Arnhold S, Skouras E, Kuerten S, Streppel M, Angelova SK, Wewetzer K, Radtke C, Dunlop SA, Angelov DN. Bone marrow-derived mesenchymal stem cell transplantation does not improve quality of muscle reinnervation or recovery of motor function after facial nerve transection in rats. Biol Chem 2008; 389:873-88. [DOI: 10.1515/bc.2008.100] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AbstractRecently, we devised and validated a novel strategy in rats to improve the outcome of facial nerve reconstruction by daily manual stimulation of the target muscles. The treatment resulted in full recovery of facial movements (whisking), which was achieved by reducing the proportion of pathologically polyinnervated motor endplates. Here, we posed whether manual stimulation could also be beneficial after a surgical procedure potentially useful for treatment of large peripheral nerve defects, i.e., entubulation of the transected facial nerve in a conduit filled with suspension of isogeneic bone marrow-derived mesenchymal stem cells (BM-MSCs) in collagen. Compared to control treatment with collagen only, entubulation with BM-MSCs failed to decrease the extent of collateral axonal branching at the lesion site and did not improve functional recovery. Post-operative manual stimulation of vibrissal muscles also failed to promote a better recovery following entubulation with BM-MSCs. We suggest that BM-MSCs promote excessive trophic support for regenerating axons which, in turn, results in excessive collateral branching at the lesion site and extensive polyinnervation of the motor endplates. Furthermore, such deleterious effects cannot be overridden by manual stimulation. We conclude that entubulation with BM-MSCs is not beneficial for facial nerve repair.
Collapse
|
27
|
Craff MN, Zeballos JL, Johnson TS, Ranka MP, Howard R, Motarjem P, Randolph MA, Winograd JM. Embryonic stem cell-derived motor neurons preserve muscle after peripheral nerve injury. Plast Reconstr Surg 2007; 119:235-245. [PMID: 17255679 DOI: 10.1097/01.prs.0000244863.71080.f0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The potential of motor neuron progenitor cell transplants to preserve muscle tissue after denervation was studied in in vivo and in vitro adult mammalian model of peripheral nerve injury. METHODS Embryonic stem cells were differentiated to induce cholinergic motor neuron progenitors. Flourescent-labeled progenitor cells were injected into the gastrocnemius muscle of Sprague-Dawley rats (n = 10) after denervation by ipilateral sciatic nerve transection. Control rats received injections of either a phosphate-buffered saline solution only (n = 12), murine embryonic fibroblast (STO) cells (n= 6), or undifferentiated embryonic stem cells (n= 6). Muscles were weighed and analyzed at 7 and 21 days using histology, histomorphometry, and immunostaining. RESULTS Seven days after progenitor cell transplant, both muscle mass and myocyte cross-sectional area were preserved, compared with control muscles, which demonstrated muscle mass reduction to 70 percent and reduction of cross-sectional area to 72 percent of normal. Fluorescent microscopy of transplanted muscles confirmed the presence of motor neuron progenitors. Presynaptic neuronal staining of the transplants overlapped with alpha-bungarotoxin-labeled muscle fibers, revealing the presence of new neuromuscular junctions. By 21 days, muscle atrophy in the experimental muscles was equal to that of controls and no transplanted cells were observed. Co-culture of the motor neuron progenitor cells and myocytes also demonstrated new neuromuscular junctions by immunofluorescence. CONCLUSIONS Transplanted motor neuron progenitors prevent muscle atrophy after denervation for a brief time. These progenitor cell transplants appear to form new neuromuscular junctions with denervated muscle fibers in vivo and with myocytes in vitro.
Collapse
Affiliation(s)
- Melody N Craff
- Boston, Mass. From the Plastic Surgery Research Laboratory, Harvard Medical School, Massachusetts General Hospital
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Bréjot T, Blanchard S, Hocquemiller M, Haase G, Liu S, Nosjean A, Heard JM, Bohl D. Forced expression of the motor neuron determinant HB9 in neural stem cells affects neurogenesis. Exp Neurol 2006; 198:167-82. [PMID: 16434037 DOI: 10.1016/j.expneurol.2005.11.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 10/12/2005] [Accepted: 11/22/2005] [Indexed: 11/25/2022]
Abstract
In contrast to mouse embryonic stem cells and in spite of overlapping gene expression profiles, neural stem cells (NSCs) isolated from the embryonic spinal cord do not respond to physiological morphogenetic stimuli provided by Sonic hedgehog and retinoic acid and do not generate motor neurons upon differentiation. Transcription factors expressed in motor neuron progenitors during embryogenesis include Pax6, Ngn2, Nkx6.1 and Olig2, whose expression precedes that of factors specifying motor neuron fate, including HB9, Islet1 and LIM3. We showed that all these factors were present in neural progenitors derived from mouse ES cells, whereas NSCs derived from the rat embryonic spinal cord expressed neither HB9 nor Islet1 and contained low levels of Nkx6.1 and LIM3. We constructed a lentivirus vector to express HB9 and GFP in NSCs and examined the consequences of HB9 expression on other transcription factors and cell differentiation. Compared to cell expressing GFP alone, NSCs expressing GFP and HB9 cycled less rapidly, downregulated Pax6 and Ngn2 mRNA levels, produced higher proportions of neurons in vitro and lower numbers of neurons after transplantation in the spinal cord of recipient rats. Oligodendrocytic and astrocytic differentiations were not affected. HB9 expressing NSCs did not express Islet1 or upregulate LIM3. They neither responded to Sonic hedgehog and retinoic acid nor produced cholinergic neurons. We concluded that forced HB9 expression affected neurogenesis but was not sufficient to confer motor neuron fate to NSCs.
Collapse
Affiliation(s)
- Thomas Bréjot
- Unité Rétrovirus et Transfert Génétique, INSERM U622, Département Neuroscience, Institut Pasteur, 28, rue du Dr. Roux, 75015 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Midha R, Munro CA, Chan S, Nitising A, Xu QG, Gordon T. Regeneration into Protected and Chronically Denervated Peripheral Nerve Stumps. Neurosurgery 2005; 57:1289-99; discussion 1289-99. [PMID: 16331178 DOI: 10.1227/01.neu.0000187480.38170.ec] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Delayed repair of peripheral nerve injuries often results in poor motor functional recovery. This may be a result of the deterioration or loss of endoneurial pathways in the distal nerve stump before motor axons can regenerate into the stump. METHODS Using the rat femoral nerve, we protected distal endoneurial pathways of the saphenous nerve with either cross-suture of the quadriceps motor nerve (Group A) or resuture of the saphenous nerve (Group B) to compare later motor regeneration into the "protected" saphenous nerve pathway to chronic denervation and "unprotected" saphenous nerve (Group C). A total of 60 rats, 20 per group, were operated on. After this protection (or lack thereof) for 8 weeks, the motor branch of the femoral nerve was cut and sutured to the distal saphenous nerve to allow motor regeneration into protected and unprotected saphenous nerve stumps. The quantitative assessment of axonal regeneration was performed after 6 weeks by use of nerve sampling for axon counts and retrogradely labeled motor neuron counts. RESULTS Significantly more myelinated axons innervated the motor (A) than the sensory (B) and no-protection (C) groups. There were significantly more retrogradely labeled femoral motor neurons in Group A than in the unprotected group (C). CONCLUSION We conclude that even 2 months of denervation of the distal nerve pathway is deleterious to regeneration and that protection of the pathway improves subsequent reinnervation and regeneration. Moreover, if the desired regeneration is motor, protection of the distal nerve pathway by a motor nerve conditions is better than a sensory nerve.
Collapse
Affiliation(s)
- Rajiv Midha
- Division of Neurosurgery, Department of Clinical Neurosciences, and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | | | | | |
Collapse
|
30
|
Desouches C, Alluin O, Mutaftschiev N, Dousset E, Magalon G, Boucraut J, Feron F, Decherchi P. La réparation nerveuse périphérique : 30 siècles de recherche. Rev Neurol (Paris) 2005; 161:1045-59. [PMID: 16288170 DOI: 10.1016/s0035-3787(05)85172-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Nerve injury compromises sensory and motor functions. Techniques of peripheral nerve repair are based on our knowledge regarding regeneration. Microsurgical techniques introduced in the late 1950s and widely developed for the past 20 years have improved repairs. However, functional recovery following a peripheral mixed nerve injury is still incomplete. STATE OF ART Good motor and sensory function after nerve injury depends on the reinnervation of the motor end plates and sensory receptors. Nerve regeneration does not begin if the cell body has not survived the initial injury or if it is unable to initiate regeneration. The regenerated axons must reach and reinnervate the appropriate target end-organs in a timely fashion. Recovery of motor function requires a critical number of motor axons reinnervating the muscle fibers. Sensory recovery is possible if the delay in reinnervation is short. Many additional factors influence the success of nerve repair or reconstruction. The timing of the repair, the level of injury, the extent of the zone of injury, the technical skill of the surgeon, and the method of repair and reconstruction contribute to the functional outcome after nerve injury. CONCLUSION This review presents the recent advances in understanding of neural regeneration and their application to the management of primary repairs and nerve gaps.
Collapse
Affiliation(s)
- C Desouches
- Service de Chirurgie de la Main, Chirurgie Plastique et Réparatrice des Membres, Assistance Publique, Hôpitaux de Marseille, Hôpital de la Conception, Marseille
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Miles GB, Yohn DC, Wichterle H, Jessell TM, Rafuse VF, Brownstone RM. Functional properties of motoneurons derived from mouse embryonic stem cells. J Neurosci 2005; 24:7848-58. [PMID: 15356197 PMCID: PMC6729934 DOI: 10.1523/jneurosci.1972-04.2004] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The capacity of embryonic stem (ES) cells to form functional motoneurons (MNs) and appropriate connections with muscle was investigated in vitro. ES cells were obtained from a transgenic mouse line in which the gene for enhanced green fluorescent protein (eGFP) is expressed under the control of the promotor of the MN specific homeobox gene Hb9. ES cells were exposed to retinoic acid (RA) and sonic hedgehog agonist (Hh-Ag1.3) to stimulate differentiation into MNs marked by expression of eGFP and the cholinergic transmitter synthetic enzyme choline acetyltransferase. Whole-cell patch-clamp recordings were made from eGFP-labeled cells to investigate the development of functional characteristics of MNs. In voltage-clamp mode, currents, including EPSCs, were recorded in response to exogenous applications of GABA, glycine, and glutamate. EGFP-labeled neurons also express voltage-activated ion channels including fast-inactivating Na(+) channels, delayed rectifier and I(A)-type K(+) channels, and Ca(2+) channels. Current-clamp recordings demonstrated that eGFP-positive neurons generate repetitive trains of action potentials and that l-type Ca(2+) channels mediate sustained depolarizations. When cocultured with a muscle cell line, clustering of acetylcholine receptors on muscle fibers adjacent to developing axons was seen. Intracellular recordings of muscle fibers adjacent to eGFP-positive axons revealed endplate potentials that increased in amplitude and frequency after glutamate application and were sensitive to TTX and curare. In summary, our findings demonstrate that MNs derived from ES cells develop appropriate transmitter receptors, intrinsic properties necessary for appropriate patterns of action potential firing and functional synapses with muscle fibers.
Collapse
Affiliation(s)
- Gareth B Miles
- Department of Anatomy and Neurobiology, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4H7
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Designing artificial microtissues by reaggregation of monodispersed primary cells, neoplastic or engineered cell lines is providing insight into cell-cell interactions and underlying regulatory networks. Recent advances in microtissue production have highlighted the potential of scaffold-free cell aggregates in maintaining tissue-specific functionality, supporting seamless integration of implants into host tissues, and providing complex feeder structures for difficult-to-differentiate cell types. Furthermore, these tissues are amenable to therapeutic and phenotype-modulating interventions using latest-generation transduction technologies. Microtissues produce therapeutic transgenes at increased levels and offer tissue-like assay environments to improve drug-function correlations in current discovery programs. Here, we outline scaffold-free microtissue design in liver, heart and cartilage, and discuss how this technology could significantly impact regenerative medicine.
Collapse
Affiliation(s)
- Jens M Kelm
- Institute of Biotechnology, Swiss Federal Institute of Technology, ETH Hoenggerberg, HPT D74, CH-8093 Zurich, Switzerland
| | | |
Collapse
|