1
|
Bonnet M, Ertlen C, Seblani M, Brezun JM, Coyle T, Cereda C, Zuccotti G, Colli M, Desouches C, Decherchi P, Carelli S, Marqueste T. Activated Human Adipose Tissue Transplantation Promotes Sensorimotor Recovery after Acute Spinal Cord Contusion in Rats. Cells 2024; 13:182. [PMID: 38247873 PMCID: PMC10814727 DOI: 10.3390/cells13020182] [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: 11/23/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Traumatic spinal cord injuries (SCIs) often result in sensory, motor, and vegetative function loss below the injury site. Although preclinical results have been promising, significant solutions for SCI patients have not been achieved through translating repair strategies to clinical trials. In this study, we investigated the effective potential of mechanically activated lipoaspirated adipose tissue when transplanted into the epicenter of a thoracic spinal contusion. Male Sprague Dawley rats were divided into three experimental groups: SHAM (uninjured and untreated), NaCl (spinal cord contusion with NaCl application), and AF (spinal cord contusion with transplanted activated human fat). Pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) were measured to assess endogenous inflammation levels 14 days after injury. Sensorimotor recovery was monitored weekly for 12 weeks, and gait and electrophysiological analyses were performed at the end of this observational period. The results indicated that AF reduced endogenous inflammation post-SCI and there was a significant improvement in sensorimotor recovery. Moreover, activated adipose tissue also reinstated the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions. This investigation highlights the efficacy of activated adipose tissue grafting in acute SCI, suggesting it is a promising therapeutic approach for spinal cord repair after traumatic contusion in humans.
Collapse
Affiliation(s)
- Maxime Bonnet
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Céline Ertlen
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Mostafa Seblani
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Jean-Michel Brezun
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Thelma Coyle
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Gianvincenzo Zuccotti
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
- Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
| | - Mattia Colli
- Podgora7 Clinic, Via Podgora 7, 20122 Milano, Italy
| | - Christophe Desouches
- Clinique Phénicia—CD Esthétique, 5 Boulevard Notre Dame, F-13006 Marseille, France
| | - Patrick Decherchi
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| | - Stephana Carelli
- Center of Functional Genomics and Rare Diseases, Department of Paediatrics, Buzzi Children’s Hospital, Via Ludovico Castelvetro 32, 20154 Milano, Italy
- Pediatric Clinical Research Center «Romeo ed Enrica Invernizzi», Department of Biomedical and Clinical Sciences, University of Milano (UNIMI), Via G.B. Grassi 74, 20157 Milan, Italy;
| | - Tanguy Marqueste
- Aix Marseille Univ, CNRS, ISM, UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe «Plasticité des Systèmes Nerveux et Musculaire» (PSNM), Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, CEDEX 09, F-13288 Marseille, France (J.-M.B.); (P.D.)
| |
Collapse
|
2
|
Gao F, Li JJ, Liu JY, Li YJ, Cong XY, Talifu Z, Zhang X. Association between brain N-acetylaspartate levels and sensory and motor dysfunction in patients who have spinal cord injury with spasticity: an observational case-control study. Neural Regen Res 2023; 18:582-586. [DOI: 10.4103/1673-5374.350216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
3
|
Zhang X, Xu L, Chen X, Zhou X, Cao L. Acacetin alleviates neuroinflammation and oxidative stress injury via the Nrf2/HO-1 pathway in a mouse model of spinal cord injury. Transl Neurosci 2022; 13:483-494. [PMID: 36590896 PMCID: PMC9773099 DOI: 10.1515/tnsci-2022-0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
Spinal cord injury (SCI) is a severe central nervous system disease, which may cause serious locomotor deficit. Acacetin is a flavone that possesses antioxidant and anti-inflammatory effects in different human diseases. The main purpose of this study was to explore whether acacetin ameliorates SCI in mice. A model of SCI was established in C57BL/6 mice. The Basso Mouse Scale (BMS) score, BMS subscore, mechanical hypersensitivity, and thermal hypersensitivity of mice were tested for determining the motor function. Immunofluorescence staining was utilized to detect NeuN, GFAP, and Iba-1 levels in spinal cord tissues. ELISA was utilized to assess the contents of proinflammatory factors such as interleukin (IL)-1β, IL-18, and tumor necrosis factor-alpha (TNF-α) in spinal cord tissues. The levels of oxidative stress markers, reactive oxygen species, thiobarbituric acid-reactive substances, superoxide dismutase, catalase, glutathione peroxidase, and glutathione were detected using their corresponding kits. Western blot was employed for estimating the levels of heme oxygenase 1 (HO-1), nuclear factor E2-related factor 2 (Nrf2), and Kelch-like ECH-associated protein 1 (Keap-1). In this study, acacetin treatment recovered the motor function in SCI mice. Acacetin improved neuron integrity and repressed glial cell activation in the spinal cord tissues of SCI mice. Furthermore, acacetin administration reduced the SCI-induced high concentrations of IL-1β, IL-18, and TNF-α, as well as inhibited oxidative stress in SCI mice. Moreover, acacetin activated HO-1/Nrf2 pathway in SCI mice. The neuroprotective effects of acacetin against SCI were reversed by Nrf2 inhibitor. Overall, acacetin alleviated neuroinflammation and oxidative stress injury by activating the Nrf2/HO-1 signaling pathway in the mouse models of SCI.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Orthopedic, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, No. 141 Tianjin Road, Huangshi 435000, Hubei, China
| | - Lijun Xu
- Department of Radiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi 435000, Hubei, China
| | - Xiang Chen
- Department of Orthopedic, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, No. 141 Tianjin Road, Huangshi 435000, Hubei, China
| | - Xianjie Zhou
- Department of Orthopedic, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, No. 141 Tianjin Road, Huangshi 435000, Hubei, China
| | - Lanhua Cao
- Department of Orthopedic, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, No. 141 Tianjin Road, Huangshi 435000, Hubei, China
| |
Collapse
|
4
|
Liu T, Zhu W, Zhang X, He C, Liu X, Xin Q, Chen K, Wang H. Recent Advances in Cell and Functional Biomaterial Treatment for Spinal Cord Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5079153. [PMID: 35978649 PMCID: PMC9377911 DOI: 10.1155/2022/5079153] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a devastating central nervous system disease caused by accidental events, resulting in loss of sensory and motor function. Considering the multiple effects of primary and secondary injuries after spinal cord injury, including oxidative stress, tissue apoptosis, inflammatory response, and neuronal autophagy, it is crucial to understand the underlying pathophysiological mechanisms, local microenvironment changes, and neural tissue functional recovery for preparing novel treatment strategies. Treatment based on cell transplantation has become the forefront of spinal cord injury therapy. The transplanted cells provide physical and nutritional support for the damaged tissue. At the same time, the implantation of biomaterials with specific biological functions at the site of the SCI has also been proved to improve the local inhibitory microenvironment and promote axonal regeneration, etc. The combined transplantation of cells and functional biomaterials for SCI treatment can result in greater neuroprotective and regenerative effects by regulating cell differentiation, enhancing cell survival, and providing physical and directional support for axon regeneration and neural circuit remodeling. This article reviews the pathophysiology of the spinal cord, changes in the microenvironment after injury, and the mechanisms and strategies for spinal cord regeneration and repair. The article will focus on summarizing and discussing the latest intervention models based on cell and functional biomaterial transplantation and the latest progress in combinational therapies in SCI repair. Finally, we propose the future prospects and challenges of current treatment regimens for SCI repair, to provide references for scientists and clinicians to seek better SCI repair strategies in the future.
Collapse
Affiliation(s)
- Tianyi Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Wenhao Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaoyu Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Chuan He
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaolong Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Qiang Xin
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Kexin Chen
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Haifeng Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
5
|
Rao Z, Lin Z, Song P, Quan D, Bai Y. Biomaterial-Based Schwann Cell Transplantation and Schwann Cell-Derived Biomaterials for Nerve Regeneration. Front Cell Neurosci 2022; 16:926222. [PMID: 35836742 PMCID: PMC9273721 DOI: 10.3389/fncel.2022.926222] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Schwann cells (SCs) dominate the regenerative behaviors after peripheral nerve injury by supporting axonal regrowth and remyelination. Previous reports also demonstrated that the existence of SCs is beneficial for nerve regeneration after traumatic injuries in central nervous system. Therefore, the transplantation of SCs/SC-like cells serves as a feasible cell therapy to reconstruct the microenvironment and promote nerve functional recovery for both peripheral and central nerve injury repair. However, direct cell transplantation often leads to low efficacy, due to injection induced cell damage and rapid loss in the circulatory system. In recent years, biomaterials have received great attention as functional carriers for effective cell transplantation. To better mimic the extracellular matrix (ECM), many biodegradable materials have been engineered with compositional and/or topological cues to maintain the biological properties of the SCs/SCs-like cells. In addition, ECM components or factors secreted by SCs also actively contribute to nerve regeneration. Such cell-free transplantation approaches may provide great promise in clinical translation. In this review, we first present the current bio-scaffolds engineered for SC transplantation and their achievement in animal models and clinical applications. To this end, we focus on the physical and biological properties of different biomaterials and highlight how these properties affect the biological behaviors of the SCs/SC-like cells. Second, the SC-derived biomaterials are also reviewed and discussed. Finally, the relationship between SCs and functional biomaterials is summarized, and the trends of their future development are predicted toward clinical applications.
Collapse
Affiliation(s)
- Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zudong Lin
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou, China
| | - Panpan Song
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Daping Quan
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Effects of mesenchymal stem cell transplantation on spinal cord injury patients. Cell Tissue Res 2022; 389:373-384. [PMID: 35697943 DOI: 10.1007/s00441-022-03648-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/02/2022] [Indexed: 11/02/2022]
Abstract
Spinal cord injury (SCI) is a traumatic injury with sensory and motor deficits that more than 1 million patients worldwide suffer from disability due to it. Many pharmacological therapies help reduce SCI-related injury and protect CNS from more damage but no current therapy could improve the axonal repair. In this regard, stem cell therapy is considered a regenerative method for SCI patient treatment. The neurotrophic and immunomodulatory factor secretion, differentiation, neuroprotecting, and remyelinating properties have made mesenchymal stem cells (MSCs) principally useful in this field. There are studies on the role of MSCs in patients suffering from SCI. However, low number of SCI patients and the lack of control groups in these studies, the cell transplantation appropriate methods, including cell source, dose, route of delivery, and transplantation timing, are various in trials. This study reviews the beneficial effects of MSC transplantation in SCI clinical studies with a special focus on the MSC properties and limitations of MSC transplantation.
Collapse
|
7
|
Preparation of Drug Sustained-Release Scaffold with De-Epithelized Human Amniotic Epithelial Cells and Thiolated Chitosan Nanocarriers and Its Repair Effect on Spinal Cord Injury. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:6294148. [PMID: 35070240 PMCID: PMC8767368 DOI: 10.1155/2022/6294148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 11/22/2022]
Abstract
The disability rate of spinal cord injury (SCI) is extremely high, and stem cell inhibition is one of the most effective schemes in treating the spinal cord, but the survival rate is extremely low after stem cell transplantation, so it cannot be widely used in clinic. Studies have revealed that loading stem cells with biological scaffolds can effectively improve the survival rate and effect after stem cell transplantation. Therefore, this research was devised to analyze the repair effect of thiolated chitosan nanocarriers scaffold carrying de-epithelized human amniotic epithelial cells (HAECs) on SCI. And we used thiolated chitosan as nanocarriers, aiming to provide a reliable theoretical basis for future clinical practice. Through experiments, we concluded that the Tarlov and BBB scores of rats with SCI were raised under the intervention of thiolated chitosan carrying HAECs, while the inflammatory factors in serum, oxidative stress reaction in spinal cord tissue, apoptosis rate of nerve cells, and autophagy protein expression were all suppressed. Thus, the thiolated chitosan carrying HAECs may be applied to treat SCI by suppressing autophagy protein expression, oxidative stress response, and release of inflammatory factors in spinal cord tissue, which may be a new clinical therapy for SCI in the future. Even though we cannot understand exactly the therapeutic mechanism of thiolated chitosan carrying HAECs for SCI, the real clinical application of thiolated chitosan carrying HAECs needs to be confirmed by human experiments.
Collapse
|
8
|
Advanced approaches to regenerate spinal cord injury: The development of cell and tissue engineering therapy and combinational treatments. Biomed Pharmacother 2021; 146:112529. [PMID: 34906773 DOI: 10.1016/j.biopha.2021.112529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) devastate event that is commonly caused by traumatic or non-traumatic events. The reinnervation of spinal cord axons is hampered through a myriad of devices counting on the damaged myelin, inflammation, glial scar, and defective inhibitory molecules. Unfortunately, an effective treatment to completely repair SCI and improve functional recovery has not been found. In this regard, strategies such as using cells, biomaterials, biomolecules, and drugs have been reported to be effective for SCI recovery. Furthermore, recent advances in combinatorial treatments, which address various aspects of SCI pathophysiology, provide optimistic outcomes for spinal cord regeneration. According to the global importance of SCI, the goal of this article review is to provide an overview of the pathophysiology of SCI, with an emphasis on the latest modes of intervention and current advanced approaches for the treatment of SCI, in conjunction with an assessment of combinatorial approaches in preclinical and clinical trials. So, this article can give scientists and clinicians' clues to help them better understand how to construct preclinical and clinical studies that could lead to a breakthrough in spinal cord regeneration.
Collapse
|
9
|
Zeraatpisheh Z, Mirzaei E, Nami M, Alipour H, Mahdavipour M, Sarkoohi P, Torabi S, Azari H, Aligholi H. Local delivery of fingolimod through PLGA nanoparticles and PuraMatrix-embedded neural precursor cells promote motor function recovery and tissue repair in spinal cord injury. Eur J Neurosci 2021; 54:5620-5637. [PMID: 34251711 DOI: 10.1111/ejn.15391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a devastating clinical problem that can lead to permanent motor dysfunction. Fingolimod (FTY720) is a sphingosine structural analogue, and recently, its therapeutic benefits in SCI have been reported. The present study aimed to evaluate the therapeutic efficacy of fingolimod-incorporated poly lactic-co-glycolic acid (PLGA) nanoparticles (nanofingolimod) delivered locally together with neural stem/progenitor cells (NS/PCs) transplantation in a mouse model of contusive acute SCI. Fingolimod was encapsulated in PLGA nanoparticles by the emulsion-evaporation method. Mouse NS/PCs were harvested and cultured from embryonic Day 14 (E14) ganglionic eminences. Induction of SCI was followed by the intrathecal delivery of nanofingolimod with and without intralesional transplantation of PuraMatrix-encapsulated NS/PCs. Functional recovery, injury size and the fate of the transplanted cells were evaluated after 28 days. The nanofingolimod particles represented spherical morphology. The entrapment efficiency determined by UV-visible spectroscopy was approximately 90%, and the drug content of fingolimod loaded nanoparticles was 13%. About 68% of encapsulated fingolimod was slowly released within 10 days. Local delivery of nanofingolimod in combination with NS/PCs transplantation led to a stronger improvement in neurological functions and minimized tissue damage. Furthermore, co-administration of nanofingolimod and NS/PCs not only increased the survival of transplanted cells but also promoted their fate towards more oligodendrocytic phenotype. Our data suggest that local release of nanofingolimod in combination with three-dimensional (3D) transplantation of NS/PCs in the acute phase of SCI could be a promising approach to restore the damaged tissues and improve neurological functions.
Collapse
Affiliation(s)
- Zahra Zeraatpisheh
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Esmaeil Mirzaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Nami
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Alipour
- Department of Tissue Engineering and Applied cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Mahdavipour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Sarkoohi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Somayyeh Torabi
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Azari
- Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Hadi Aligholi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Sykova E, Cizkova D, Kubinova S. Mesenchymal Stem Cells in Treatment of Spinal Cord Injury and Amyotrophic Lateral Sclerosis. Front Cell Dev Biol 2021; 9:695900. [PMID: 34295897 PMCID: PMC8290345 DOI: 10.3389/fcell.2021.695900] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/31/2021] [Indexed: 01/01/2023] Open
Abstract
Preclinical and clinical studies with various stem cells, their secretomes, and extracellular vesicles (EVs) indicate their use as a promising strategy for the treatment of various diseases and tissue defects, including neurodegenerative diseases such as spinal cord injury (SCI) and amyotrophic lateral sclerosis (ALS). Autologous and allogenic mesenchymal stem cells (MSCs) are so far the best candidates for use in regenerative medicine. Here we review the effects of the implantation of MSCs (progenitors of mesodermal origin) in animal models of SCI and ALS and in clinical studies. MSCs possess multilineage differentiation potential and are easily expandable in vitro. These cells, obtained from bone marrow (BM), adipose tissue, Wharton jelly, or even other tissues, have immunomodulatory and paracrine potential, releasing a number of cytokines and factors which inhibit the proliferation of T cells, B cells, and natural killer cells and modify dendritic cell activity. They are hypoimmunogenic, migrate toward lesion sites, induce better regeneration, preserve perineuronal nets, and stimulate neural plasticity. There is a wide use of MSC systemic application or MSCs seeded on scaffolds and tissue bridges made from various synthetic and natural biomaterials, including human decellularized extracellular matrix (ECM) or nanofibers. The positive effects of MSC implantation have been recorded in animals with SCI lesions and ALS. Moreover, promising effects of autologous as well as allogenic MSCs for the treatment of SCI and ALS were demonstrated in recent clinical studies.
Collapse
Affiliation(s)
- Eva Sykova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Dasa Cizkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia.,Centre for Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia
| | - Sarka Kubinova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
11
|
Abbas WA, Ibrahim ME, El-Naggar M, Abass WA, Abdullah IH, Awad BI, Allam NK. Recent Advances in the Regenerative Approaches for Traumatic Spinal Cord Injury: Materials Perspective. ACS Biomater Sci Eng 2020; 6:6490-6509. [PMID: 33320628 DOI: 10.1021/acsbiomaterials.0c01074] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) is a devastating health condition that may lead to permanent disabilities and death. Understanding the pathophysiological perspectives of traumatic SCI is essential to define mechanisms that can help in designing recovery strategies. Since central nervous system tissues are notorious for their deficient ability to heal, efforts have been made to identify solutions to aid in restoration of the spinal cord tissues and thus its function. The two main approaches proposed to address this issue are neuroprotection and neuro-regeneration. Neuroprotection involves administering drugs to restore the injured microenvironment to normal after SCI. As for the neuro-regeneration approach, it focuses on axonal sprouting for functional recovery of the injured neural tissues and damaged axons. Despite the progress made in the field, neural regeneration treatment after SCI is still unsatisfactory owing to the disorganized way of axonal growth and extension. Nanomedicine and tissue engineering are considered promising therapeutic approaches that enhance axonal growth and directionality through implanting or injecting of the biomaterial scaffolds. One of these recent approaches is nanofibrous scaffolds that are used to provide physical support to maintain directional axonal growth in the lesion site. Furthermore, these preferable tissue-engineered substrates can afford axonal regeneration by mimicking the extracellular matrix of the neural tissues in terms of biological, chemical, and architectural characteristics. In this review, we discuss the regenerative approach using nanofibrous scaffolds with a focus on their fabrication methods and their properties that define their functionality performed to heal the neural tissue efficiently.
Collapse
Affiliation(s)
- Walaa A Abbas
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Maha E Ibrahim
- Department of Physical Medicine, Rheumatology and Rehabilitation, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Manar El-Naggar
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Wessam A Abass
- Center of Sustainable Development, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Ibrahim H Abdullah
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Basem I Awad
- Mansoura Experimental Research Center (MERC), Department of Neurological Surgery, School of Medicine, Mansoura University, Mansoura, Egypt
| | - Nageh K Allam
- Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
12
|
Spinal Reflex Recovery after Dorsal Rhizotomy and Repair with Platelet-Rich Plasma (PRP) Gel Combined with Bioengineered Human Embryonic Stem Cells (hESCs). Stem Cells Int 2020; 2020:8834360. [PMID: 33178285 PMCID: PMC7647752 DOI: 10.1155/2020/8834360] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/20/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Dorsal root rhizotomy (DRZ) is currently considered an untreatable injury, resulting in the loss of sensitive function and usually leading to neuropathic pain. In this context, we recently proposed a new surgical approach to treat DRZ that uses platelet-rich plasma (PRP) gel to restore the spinal reflex. Success was correlated with the reentry of primary afferents into the spinal cord. Here, aiming to enhance previous results, cell therapy with bioengineered human embryonic stem cells (hESCs) to overexpress fibroblast growth factor 2 (FGF2) was combined with PRP. For these experiments, adult female rats were submitted to a unilateral rhizotomy of the lumbar spinal dorsal roots, which was followed by root repair with PRP gel with or without bioengineered hESCs. One week after DRZ, the spinal cords were processed to evaluate changes in the glial response (GFAP and Iba-1) and excitatory synaptic circuits (VGLUT1) by immunofluorescence. Eight weeks postsurgery, the lumbar intumescences were processed for analysis of the repaired microenvironment by transmission electron microscopy. Spinal reflex recovery was evaluated by the electronic Von Frey method for eight weeks. The transcript levels for human FGF2 were over 37-fold higher in the induced hESCs than in the noninduced and the wildtype counterparts. Altogether, the results indicate that the combination of hESCs with PRP gel promoted substantial and prominent axonal regeneration processes after DRZ. Thus, the repair of dorsal roots, if done appropriately, may be considered an approach to regain sensory-motor function after dorsal root axotomy.
Collapse
|
13
|
Vikartovska Z, Kuricova M, Farbakova J, Liptak T, Mudronova D, Humenik F, Madari A, Maloveska M, Sykova E, Cizkova D. Stem Cell Conditioned Medium Treatment for Canine Spinal Cord Injury: Pilot Feasibility Study. Int J Mol Sci 2020; 21:ijms21145129. [PMID: 32698543 PMCID: PMC7404210 DOI: 10.3390/ijms21145129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) involves nerve damage and often leads to motor, sensory and autonomic dysfunctions. In the present study, we have designed a clinical protocol to assess the feasibility of systemic delivery of allogenic canine bone marrow tissue-derived mesenchymal stem cell conditioned medium (BMMSC CM) to dogs with SCI. Four client-owned dogs with chronic SCI lasting more than six months underwent neurological and clinical evaluation, MRI imaging and blood tests before being enrolled in this study. All dogs received four intravenous infusions with canine allogenic BMMSC CM within one month. Between the infusions the dogs received comprehensive physiotherapy, which continued for three additional months. No adverse effects or complications were observed during the one, three and six months follow-up periods. Neither blood chemistry panel nor hematology profile showed any significant changes. All dogs were clinically improved as assessed using Olby locomotor scales after one, three and six months of BMMSC CM treatment. Furthermore, goniometric measurements revealed partial improvement in the range of joint motion. Bladder function improved in two disabled dogs. We conclude that multiple delivery of allogenic cell-derived conditioned medium to dogs with chronic SCI is feasible, and it might be clinically beneficial in combination with physiotherapy.
Collapse
Affiliation(s)
- Zuzana Vikartovska
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Maria Kuricova
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Jana Farbakova
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Tomas Liptak
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Dagmar Mudronova
- Department of Microbiology and Immunology, Institute of Immunology, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia;
| | - Filip Humenik
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
| | - Aladar Madari
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Marcela Maloveska
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
| | - Eva Sykova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia;
| | - Dasa Cizkova
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia;
- Correspondence:
| |
Collapse
|
14
|
Delivery of Antisense Oligonucleotides Mediated by a Hydrogel System: In Vitro and In Vivo Application in the Context of Spinal Cord Injury. Methods Mol Biol 2020; 2036:205-219. [PMID: 31410799 DOI: 10.1007/978-1-4939-9670-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Biomaterials-based hydrogels are attractive drug-eluting vehicles in the context of RNA therapeutics, such as those utilizing antisense oligonucleotide or RNA interference based drugs, as they can potentially reduce systemic toxicity and enhance in vivo efficacy by increasing in situ concentrations. Here we describe the preparation of antisense oligonucleotide-loaded fibrin hydrogels exploring their applications in the context of the nervous system utilizing an organotypic dorsal root ganglion explant in vitro system and an in vivo model of spinal cord injury.
Collapse
|
15
|
Bydon M, Dietz AB, Goncalves S, Moinuddin FM, Alvi MA, Goyal A, Yolcu Y, Hunt CL, Garlanger KL, Del Fabro AS, Reeves RK, Terzic A, Windebank AJ, Qu W. CELLTOP Clinical Trial: First Report From a Phase 1 Trial of Autologous Adipose Tissue-Derived Mesenchymal Stem Cells in the Treatment of Paralysis Due to Traumatic Spinal Cord Injury. Mayo Clin Proc 2020; 95:406-414. [PMID: 31785831 DOI: 10.1016/j.mayocp.2019.10.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/30/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition with limited pharmacological treatment options to restore function. Regenerative approaches have recently attracted interest as an adjuvant to current standard of care. Adipose tissue-derived (AD) mesenchymal stem cells (MSCs) represent a readily accessible cell source with high proliferative capacity. The CELLTOP study, an ongoing multidisciplinary phase 1 clinical trial conducted at Mayo Clinic (ClinicalTrials.gov Identifier: NCT03308565), is investigating the safety and efficacy of intrathecal autologous AD-MSCs in patients with blunt, traumatic SCI. In this initial report, we describe the outcome of the first treated patient, a 53-year-old survivor of a surfing accident who sustained a high cervical American Spinal Injury Association Impairment Scale grade A SCI with subsequent neurologic improvement that plateaued within 6 months following injury. Although he improved to an American Spinal Injury Association grade C impairement classification, the individual continued to be wheelchair bound and severely debilitated. After study enrollment, an adipose tissue biopsy was performed and MSCs were isolated, expanded, and cryopreserved. Per protocol, the patient received an intrathecal injection of 100 million autologous AD-MSCs infused after a standard lumbar puncture at the L3-4 level 11 months after the injury. The patient tolerated the procedure well and did not experience any severe adverse events. Clinical signs of efficacy were observed at 3, 6, 12, and 18 months following the injection in both motor and sensory scores based on International Standards for Neurological Classification of Spinal Cord Injury. Thus, in this treated individual with SCI, intrathecal administration of AD-MSCs was feasible and safe and suggested meaningful signs of improved, rather than stabilized, neurologic status warranting further clinical evaluation.
Collapse
Affiliation(s)
- Mohamad Bydon
- Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN.
| | - Allan B Dietz
- Division of Transfusion Medicine, Mayo Clinic, Rochester, MN
| | - Sandy Goncalves
- Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - F M Moinuddin
- Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - Mohammed Ali Alvi
- Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - Anshit Goyal
- Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - Yagiz Yolcu
- Neuro-Informatics Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN
| | - Christine L Hunt
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, Mayo Clinic, Rochester, MN
| | - Kristin L Garlanger
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Anna S Del Fabro
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Ronald K Reeves
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Andre Terzic
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN; Department of Clinical Genomics, Mayo Clinic, Rochester, MN
| | | | - Wenchun Qu
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN.
| |
Collapse
|
16
|
Ma D, Zhao Y, Huang L, Xiao Z, Chen B, Shi Y, Shen H, Dai J. A novel hydrogel-based treatment for complete transection spinal cord injury repair is driven by microglia/macrophages repopulation. Biomaterials 2020; 237:119830. [PMID: 32036301 DOI: 10.1016/j.biomaterials.2020.119830] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 12/28/2019] [Accepted: 01/25/2020] [Indexed: 12/17/2022]
Abstract
Microglia/macrophage mediated-inflammation, a main contributor to the microenvironment after spinal cord injury (SCI), persists for a long period of time and affects SCI repair. However, the effects of microglia/macrophage mediated-inflammation on neurogenic differentiation of endogenous neural stem/progenitor cells (NSPCs) are not well understood. In this study, to attenuate activated microglia/macrophage mediated-inflammation in the spinal cord of complete transection SCI mice, a combination of photo-crosslinked hydrogel transplantation and CSF1R inhibitor (PLX3397) treatment was used to replace the prolonged, activated microglia/macrophages via cell depletion and repopulation. This combined treatment in SCI mice produced a significant reduction in CD68-positive reactive microglia/macrophages and mRNA levels of pro-inflammatory factors, and a substantial increase in the number of Tuj1-positive neurons in the lesion area compared with single treatment methods. Moreover, most of the newborn Tuj1-positive neurons were confirmed to be generated from endogenous NSPCs using a genetic fate mapping mouse line (Nestin-CreERT2; LSL-tdTomato) that can label and trace NSPC marker-nestin expressing cells and their progenies. Collectively, our findings show that the combined treatment method for inhibiting microglia/macrophage mediated-inflammation promotes endogenous NSPC neurogenesis and improves functional recovery, which provides a promising therapeutic strategy for complete transection SCI.
Collapse
Affiliation(s)
- Dezun Ma
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Lei Huang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, PR China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, PR China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Beijing, 100101, PR China.
| |
Collapse
|
17
|
The Effect of Wharton Jelly-Derived Mesenchymal Stromal Cells and Their Conditioned Media in the Treatment of a Rat Spinal Cord Injury. Int J Mol Sci 2019; 20:ijms20184516. [PMID: 31547264 PMCID: PMC6770545 DOI: 10.3390/ijms20184516] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
The transplantation of Wharton’s jelly derived mesenchymal stromal cells (WJ-MSCs) possesses therapeutic potential for the treatment of a spinal cord injury (SCI). Generally, the main effect of MSCs is mediated by their paracrine potential. Therefore, application of WJ-MSC derived conditioned media (CM) is an acknowledged approach for how to bypass the limited survival of transplanted cells. In this study, we compared the effect of human WJ-MSCs and their CM in the treatment of SCI in rats. WJ-MSCs and their CM were intrathecally transplanted in the three consecutive weeks following the induction of a balloon compression lesion. Behavioral analyses were carried out up to 9 weeks after the SCI and revealed significant improvement after the treatment with WJ-MSCs and CM, compared to the saline control. Both WJ-MSCs and CM treatment resulted in a higher amount of spared gray and white matter and enhanced expression of genes related to axonal growth. However, only the CM treatment further improved axonal sprouting and reduced the number of reactive astrocytes in the lesion area. On the other hand, WJ-MSCs enhanced the expression of inflammatory and chemotactic markers in plasma, which indicates a systemic immunological response to xenogeneic cell transplantation. Our results confirmed that WJ-MSC derived CM offer an alternative to direct stem cell transplantation for the treatment of SCI.
Collapse
|
18
|
Biomaterials and Magnetic Stem Cell Delivery in the Treatment of Spinal Cord Injury. Neurochem Res 2019; 45:171-179. [DOI: 10.1007/s11064-019-02808-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 12/23/2022]
|
19
|
Ko CC, Tu TH, Wu JC, Huang WC, Cheng H. Acidic Fibroblast Growth Factor in Spinal Cord Injury. Neurospine 2019; 16:728-738. [PMID: 30653905 PMCID: PMC6944993 DOI: 10.14245/ns.1836216.108] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 12/26/2018] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI), with an incidence rate of 246 per million person-years among adults in Taiwan, remains a devastating disease in the modern day. Elderly men with lower socioeconomic status have an even higher risk for SCI. Despite advances made in medicine and technology to date, there are few effective treatments for SCI due to limitations in the regenerative capacity of the adult central nervous system. Experiments and clinical trials have explored neuro-regeneration in human SCI, encompassing cell- and molecule-based therapies. Furthermore, strategies have aimed at restoring connections, including autologous peripheral nerve grafts and biomaterial scaffolds that theoretically promote axonal growth. Most molecule-based therapies target the modulation of inhibitory molecules to promote axonal growth, degrade glial scarring obstacles, and stimulate intrinsic regenerative capacity. Among them, acidic fibroblast growth factor (aFGF) has been investigated for nerve repair; it is mitogenic and pluripotent in nature and could enhance axonal growth and mitigate glial scarring. For more than 2 decades, the authors have conducted multiple trials, including human and animal experiments, using aFGF to repair nerve injuries, including central and peripheral nerves. In these trials, aFGF has shown promise for neural regeneration, and in the future, more trials and applications should investigate aFGF as a neurotrophic factor. Focusing on aFGF, the current review aimed to summarize the historical evolution of the utilization of aFGF in SCI and nerve injuries, to present applications and trials, to summarize briefly its possible mechanisms, and to provide future perspectives.
Collapse
Affiliation(s)
- Chin-Chu Ko
- Jhong Jheng Spine & Orthopedic Hospital, Kaohsiung, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tsung-Hsi Tu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Jau-Ching Wu
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Cheng Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Henrich Cheng
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
20
|
Yang P, Chen A, Qin Y, Yin J, Cai X, Fan YJ, Li L, Huang HY. Buyang huanwu decoction combined with BMSCs transplantation promotes recovery after spinal cord injury by rescuing axotomized red nucleus neurons. JOURNAL OF ETHNOPHARMACOLOGY 2019; 228:123-131. [PMID: 30266421 DOI: 10.1016/j.jep.2018.09.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/09/2018] [Accepted: 09/24/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Buyang huanwu decoction (BYHWD) is a classic recipe in traditional Chinese medicine (TCM) to supplement Qi and activate blood. It has been used to recover the neural function after the injury of central nervous system for hundreds of years in China. AIM OF THE STUDY This study investigated whether Buyang huanwu decoction (BYHWD) combined with bone marrow mesenchymal stem cells (BMSCs) transplantation had synergistic effect on neuroprotection of red nucleus neurons after spinal cord injury (SCI). MATERIALS AND METHODS Rubrospinal tract (RST) transection model was established and BMSCs were collected. The forelimb locomotor function was recorded using inclined plate test and spontaneous vertical exploration. cAMP level in red nucleus was detected with Enzyme-linked immunosorbent assay (ELISA). Morphology and number of red nucleus neurons was observed using Nissl's staining. Expression of cAMP-response element binding protein (CREB), ras homolog gene family member A (RhoA) and nerve growth factor (NGF) in red nucleus was detected using immunohistochemistry, qRT-PCR and Western-blotting. RESULTS The combination of BYHWD and BMSCs transplantation could improve the forelimb locomotor function significantly and give the red nucleus somas a better protection. Meanwhile, cAMP level, CREB and NGF increased, while RhoA decreased remarkably in the BYHWD+BMSCs group. CONCLUSIONS BYHWD combined with BMSCs transplantation had synergistic effect on neuroprotection of red nucleus neurons after SCI; the mechanism may be related to up-regulating cAMP level, activating the cAMP/CREB/RhoA signaling pathway, and promoting expression of NGF.
Collapse
Affiliation(s)
- Ping Yang
- Department of Psychiatry, Hunan Brain Hospital, NO.427, Middle Furong Road, Changsha, Hunan Province 410007, China
| | - An Chen
- Department of Anatomy, Hunan University of Chinese Medicine, NO.300, Xue shi Road, Changsha, Hunan Province 410208, China
| | - You Qin
- Institute of Chinese Materia Medica, Hunan Academy of Chinese Medicine, NO. 8, Yuehua Road, Changsha, Hunan Province 410013, China
| | - Jian Yin
- Department of Anatomy, Hunan University of Chinese Medicine, NO.300, Xue shi Road, Changsha, Hunan Province 410208, China
| | - Xiong Cai
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, NO.300, Xue shi Road, Changsha, Hunan Province 410208, China
| | - Yu-Jie Fan
- Department of Psychiatry, Hunan Brain Hospital, NO.427, Middle Furong Road, Changsha, Hunan Province 410007, China
| | - Liang Li
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, NO.300, Xue shi Road, Changsha, Hunan Province 410208, China.
| | - Hui-Yong Huang
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, NO.300, Xue shi Road, Changsha, Hunan Province 410208, China.
| |
Collapse
|
21
|
3D MALDI mass spectrometry imaging reveals specific localization of long-chain acylcarnitines within a 10-day time window of spinal cord injury. Sci Rep 2018; 8:16083. [PMID: 30382158 PMCID: PMC6208337 DOI: 10.1038/s41598-018-34518-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/19/2018] [Indexed: 12/14/2022] Open
Abstract
We report, for the first time, the detection and specific localization of long-chain acylcarnitines (LC ACs) along the lesion margins in an experimental model of spinal cord injury (SCI) using 3D mass spectrometry imaging (MSI). Acylcarnitines palmitoylcarnitine (AC(16:0)), palmitoleoylcarnitine (AC(16:1)), elaidic carnitine (AC(18:1)) and tetradecanoylcarnitine (AC(14:1)) were detected as early as 3 days post injury, and were present along the lesion margins 7 and 10 days after SCI induced by balloon compression technique in the rat. 3D MSI revealed the heterogeneous distribution of these lipids across the injured spinal cord, appearing well-defined at the lesion margins rostral to the lesion center, and becoming widespread and less confined to the margins at the region located caudally. The assigned acylcarnitines co-localize with resident microglia/macrophages detected along the lesion margins by immunofluorescence. Given the reported pro-inflammatory role of these acylcarnitines, their specific spatial localization along the lesion margin could hint at their potential pathophysiological roles in the progression of SCI.
Collapse
|
22
|
Mauri E, Sacchetti A, Vicario N, Peruzzotti-Jametti L, Rossi F, Pluchino S. Evaluation of RGD functionalization in hybrid hydrogels as 3D neural stem cell culture systems. Biomater Sci 2018; 6:501-510. [PMID: 29368775 DOI: 10.1039/c7bm01056g] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The use of neural stem cells (NSCs) in cell therapy has become a powerful tool used for the treatment of central nervous system diseases, including traumatic brain and spinal cord injuries. However, a significant drawback is related to the limited viability after transplantation in situ. The design of three-dimensional (3D) scaffolds that are capable of resembling the architecture and physico-chemical features of an extracellular environment could be a suitable approach to improve cell survival and preserve their cellular active phase over time. In this study, we investigated NSC adhesion and proliferation in hydrogel systems. In particular, we evaluated the effect of RGD binding domains on cell fate within the polymeric scaffold. The introduction of a tripeptide via hydrogel chemical functionalization improved the percentage of proliferating cells until 8 days after seeding when compared to the unmodified scaffold. The beneficial effects of this 3D culture system was further evident when compared to a NSC monolayer (2D) culture, resulting in an approximately 40% increase in cells in the active phases at 4 and 8 days, and maintained a difference of 25% until 21 days after seeding.
Collapse
Affiliation(s)
- Emanuele Mauri
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
23
|
Moreno PMD, Ferreira AR, Salvador D, Rodrigues MT, Torrado M, Carvalho ED, Tedebark U, Sousa MM, Amaral IF, Wengel J, Pêgo AP. Hydrogel-Assisted Antisense LNA Gapmer Delivery for In Situ Gene Silencing in Spinal Cord Injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:393-406. [PMID: 29858074 PMCID: PMC5992461 DOI: 10.1016/j.omtn.2018.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 03/04/2018] [Accepted: 03/15/2018] [Indexed: 12/16/2022]
Abstract
After spinal cord injury (SCI), nerve regeneration is severely hampered due to the establishment of a highly inhibitory microenvironment at the injury site, through the contribution of multiple factors. The potential of antisense oligonucleotides (AONs) to modify gene expression at different levels, allowing the regulation of cell survival and cell function, together with the availability of chemically modified nucleic acids with favorable biopharmaceutical properties, make AONs an attractive tool for novel SCI therapy developments. In this work, we explored the potential of locked nucleic acid (LNA)-modified AON gapmers in combination with a fibrin hydrogel bridging material to induce gene silencing in situ at a SCI lesion site. LNA gapmers were effectively developed against two promising gene targets aiming at enhancing axonal regeneration-RhoA and GSK3β. The fibrin-matrix-assisted AON delivery system mediated potent RNA knockdown in vitro in a dorsal root ganglion explant culture system and in vivo at a SCI lesion site, achieving around 75% downregulation 5 days after hydrogel injection. Our results show that local implantation of a AON-gapmer-loaded hydrogel matrix mediated efficient gene silencing in the lesioned spinal cord and is an innovative platform that can potentially combine gene regulation with regenerative permissive substrates aiming at SCI therapeutics and nerve regeneration.
Collapse
Affiliation(s)
- Pedro M D Moreno
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ana R Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Daniela Salvador
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Maria T Rodrigues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Marília Torrado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Eva D Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Ulf Tedebark
- GE Healthcare Bio-Sciences AB, 75184 Uppsala, Sweden; SynMer AB, 17568 Järfälla, Sweden
| | - Mónica M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Nerve Regeneration Group, Universidade do Porto, 4200-135 Porto, Portugal
| | - Isabel F Amaral
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Jesper Wengel
- Nucleic Acid Center, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
| | - Ana P Pêgo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; Faculdade de Engenharia da Universidade do Porto, 4200-465 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
24
|
Cizkova D, Cubinkova V, Smolek T, Murgoci AN, Danko J, Vdoviakova K, Humenik F, Cizek M, Quanico J, Fournier I, Salzet M. Localized Intrathecal Delivery of Mesenchymal Stromal Cells Conditioned Medium Improves Functional Recovery in a Rat Model of Spinal Cord Injury. Int J Mol Sci 2018; 19:ijms19030870. [PMID: 29543759 PMCID: PMC5877731 DOI: 10.3390/ijms19030870] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/04/2018] [Accepted: 03/09/2018] [Indexed: 12/16/2022] Open
Abstract
It was recently shown that the conditioned medium (CM) of mesenchymal stem cells can enhance viability of neural and glial cell populations. In the present study, we have investigated a cell-free approach via CM from rat bone marrow stromal cells (MScCM) applied intrathecally (IT) for spinal cord injury (SCI) recovery in adult rats. Functional in vitro test on dorsal root ganglion (DRG) primary cultures confirmed biological properties of collected MScCM for production of neurosphere-like structures and axon outgrowth. Afterwards, rats underwent SCI and were treated with IT delivery of MScCM or vehicle at postsurgical Days 1, 5, 9, and 13, and left to survive 10 weeks. Rats that received MScCM showed significantly higher motor function recovery, increase in spared spinal cord tissue, enhanced GAP-43 expression and attenuated inflammation in comparison with vehicle-treated rats. Spared tissue around the lesion site was infiltrated with GAP-43-labeled axons at four weeks that gradually decreased at 10 weeks. Finally, a cytokine array performed on spinal cord extracts after MScCM treatment revealed decreased levels of IL-2, IL-6 and TNFα when compared to vehicle group. In conclusion, our results suggest that molecular cocktail found in MScCM is favorable for final neuroregeneration after SCI.
Collapse
Affiliation(s)
- Dasa Cizkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Veronika Cubinkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
| | - Adriana-Natalia Murgoci
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Jan Danko
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Katarina Vdoviakova
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Filip Humenik
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Milan Cizek
- Department of Epizootology and Parasitology, University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 041 81 Košice, Slovakia.
| | - Jusal Quanico
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Isabelle Fournier
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| | - Michel Salzet
- Université de Lille, Inserm, U-1192-Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France.
| |
Collapse
|
25
|
Zaviskova K, Tukmachev D, Dubisova J, Vackova I, Hejcl A, Bystronova J, Pravda M, Scigalkova I, Sulakova R, Velebny V, Wolfova L, Kubinova S. Injectable hydroxyphenyl derivative of hyaluronic acid hydrogel modified with RGD as scaffold for spinal cord injury repair. J Biomed Mater Res A 2018; 106:1129-1140. [PMID: 29266693 DOI: 10.1002/jbm.a.36311] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/09/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022]
Abstract
Hydrogel scaffolds which bridge the lesion, together with stem cell therapy represent a promising approach for spinal cord injury (SCI) repair. In this study, a hydroxyphenyl derivative of hyaluronic acid (HA-PH) was modified with the integrin-binding peptide arginine-glycine-aspartic acid (RGD), and enzymatically crosslinked to obtain a soft injectable hydrogel. Moreover, addition of fibrinogen was used to enhance proliferation of human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSCs) on HA-PH-RGD hydrogel. The neuroregenerative potential of HA-PH-RGD hydrogel was evaluated in vivo in acute and subacute models of SCI. Both HA-PH-RGD hydrogel injection and implantation into the acute spinal cord hemisection cavity resulted in the same axonal and blood vessel density in the lesion area after 2 and 8 weeks. HA-PH-RGD hydrogel alone or combined with fibrinogen (HA-PH-RGD/F) and seeded with hWJ-MSCs was then injected into subacute SCI and evaluated after 8 weeks using behavioural, histological and gene expression analysis. A subacute injection of both HA-PH-RGD and HA-PH-RGD/F hydrogels similarly promoted axonal ingrowth into the lesion and this effect was further enhanced when the HA-PH-RGD/F was combined with hWJ-MSCs. On the other hand, no effect was found on locomotor recovery or the blood vessel ingrowth and density of glial scar around the lesion. In conclusion, we have developed and characterized injectable HA-PH-RGD based hydrogel, which represents a suitable material for further combinatorial therapies in neural tissue engineering. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 1129-1140, 2018.
Collapse
Affiliation(s)
- Kristyna Zaviskova
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic.,2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - Dmitry Tukmachev
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic.,2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - Jana Dubisova
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic.,2nd Medical Faculty, Charles University, Prague, Czech Republic
| | - Irena Vackova
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ales Hejcl
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Julie Bystronova
- Department of Tissue Engineering, Contipro a.s., Dolni Dobrouc, Czech Republic
| | - Martin Pravda
- Department of Tissue Engineering, Contipro a.s., Dolni Dobrouc, Czech Republic
| | - Ivana Scigalkova
- Department of Tissue Engineering, Contipro a.s., Dolni Dobrouc, Czech Republic
| | - Romana Sulakova
- Department of Tissue Engineering, Contipro a.s., Dolni Dobrouc, Czech Republic
| | - Vladimir Velebny
- Department of Tissue Engineering, Contipro a.s., Dolni Dobrouc, Czech Republic
| | - Lucie Wolfova
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic.,Department of Tissue Engineering, Contipro a.s., Dolni Dobrouc, Czech Republic
| | - Sarka Kubinova
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
26
|
Collagen Implant and Mononuclear Cells of Umbilical Blood Allow the Restore of Movements of Hind Limbs after Removing the Site of Spinal Cord. Bull Exp Biol Med 2018; 164:390-393. [DOI: 10.1007/s10517-018-3996-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Indexed: 10/18/2022]
|
27
|
Laminin-derived Ile-Lys-Val-ala-Val: a promising bioactive peptide in neural tissue engineering in traumatic brain injury. Cell Tissue Res 2017; 371:223-236. [PMID: 29082446 DOI: 10.1007/s00441-017-2717-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/10/2017] [Indexed: 01/09/2023]
Abstract
The adult brain has a very limited regeneration capacity and there is no effective treatment currently available for brain injury. Neuroprotective drugs aim to reduce the intensity of cell degeneration but do not trigger tissue regeneration. Cell replacement therapy is a novel strategy to overcome brain injury-induced disability. To enhance cell viability and neuronal differentiation, developing bioactive scaffolds combined with stem cells for transplantation is a crucial approach in brain tissue engineering. Cell interactions with the extracellular matrix (ECM) play a vital role in neuronal cell survival, neurite outgrowth, attachment, migration, differentiation, and proliferation. Thus, appropriate cell-ECM interactions are essential when designing and modifying scaffolds for application in neural tissue engineering. To improve cell-ECM interactions, scaffolds can be modified with bioactive peptides. Here, we discuss the characteristic features of laminin-derived Ile-Lys-Val-Ala-Val (IKVAV) sequence as a bio-functional motif in scaffolds and the behavior of stem cells in scaffolds conjugated with the IKVAV peptide. The incorporation of this bioactive peptide in nanofiber scaffolds markedly improves stem cell behavior and may be a potential method for cell replacement therapy in traumatic brain injury.
Collapse
|
28
|
Pluripotent Stem Cells and Other Innovative Strategies for the Treatment of Ocular Surface Diseases. Stem Cell Rev Rep 2017; 12:171-8. [PMID: 26779895 DOI: 10.1007/s12015-016-9643-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cornea provides two thirds of the refractive power of the eye and protection against insults such as infection and injury. The outermost tissue of the cornea is renewed by stem cells located in the limbus. Depletion or destruction of these stem cells may lead to blinding limbal stem cell deficiency (LSCD) that concerns millions of patients around the world. Innovative strategies based on adult stem cell therapies have been developed in the recent years but they are still facing numerous unresolved issues, and the long term results can be deceiving. Today there is a clear need to improve these therapies, and/or to develop new approaches for the treatment of LSCD. Here, we review the current cell-based therapies used for the treatment of ocular diseases, and discuss the potential of pluripotent stem cells (embryonic and induced pluripotent stem cells) in corneal repair. As the secretion of paracrine factors is known to have a crucial role in maintaining stem cell homeostasis and in wound repair, we also consider the therapeutic potential of a promising novel pathway, the exosomes. Exosomes are nano-sized vesicles that have the ability to transfer RNAs and proteins to recipient cells, and several studies demonstrated their role in cell protection and wound healing. Exosomes could circumvent the hurdles of stem-cell based approaches, and they could become a strong candidate as an alternative therapy for ocular surface diseases.
Collapse
|
29
|
Chedly J, Soares S, Montembault A, von Boxberg Y, Veron-Ravaille M, Mouffle C, Benassy MN, Taxi J, David L, Nothias F. Physical chitosan microhydrogels as scaffolds for spinal cord injury restoration and axon regeneration. Biomaterials 2017; 138:91-107. [DOI: 10.1016/j.biomaterials.2017.05.024] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/04/2023]
|
30
|
Silva J, Bento AR, Barros D, Laundos TL, Sousa SR, Quelhas P, Sousa MM, Pêgo AP, Amaral IF. Fibrin functionalization with synthetic adhesive ligands interacting with α6β1 integrin receptor enhance neurite outgrowth of embryonic stem cell-derived neural stem/progenitors. Acta Biomater 2017; 59:243-256. [PMID: 28694236 DOI: 10.1016/j.actbio.2017.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 12/25/2022]
Abstract
To enhance fibrin hydrogel affinity towards pluripotent stem cell-derived neural stem/progenitor cells (NSPCs) and its capacity to support NSPC migration and neurite extension, we explored the tethering of synthetic peptides engaging integrin α6β1, a cell receptor enriched in NSPCs. Six α6β1 integrin ligands were tested for their ability to support integrin α6β1-mediated adhesion of embryonic stem cell-derived NSPCs (ES-NSPs) and sustain ES-NSPC viability, migration, and neuronal differentiation. Due to their better performance, peptides T1, HYD1, and A5G81 were immobilized into fibrin and functionalized gels characterized in terms of peptide binding efficiency, structure and viscoelastic properties. Tethering of T1 or HYD1 successfully enhanced cell outgrowth from ES-NSPC neurospheres (up to 2.4-fold increase), which exhibited a biphasic response to peptide concentration. Inhibition assays evidenced the involvement of α6β1 and α3β1 integrins in mediating radial outgrowth on T1-/HYD1-functionalized gels. Fibrin functionalization also promoted neurite extension of single ES-NSPCs in fibrin, without affecting cell proliferation and neuronal differentiation. Finally, HYD1-functionalized gels were found to provide a permissive environment for axonal regeneration, leading up to a 2.0-fold increase in neurite extension from rat dorsal root ganglia explants as compared to unmodified fibrin, and to significant improved locomotor function after spinal cord injury (complete transection), along with a trend toward a higher area positive for growth associated protein 43 (marker for axonal growth cone formation). Our results suggest that conjugation of α6β1 integrin-binding motifs is of interest to increase the biofunctionality of hydrogels used in 3D platforms for ES-NSPC culture and potentially, in matrix-assisted ES-NSPC transplantation. STATEMENT OF SIGNIFICANCE Impact statement: The transplantation of NSPCs derived from pluripotent stem cells holds much promise for the treatment of central nervous system disorders. Moreover, the combinatorial use of biodegradable hydrogels with NSPCs was shown to contribute to the establishment of a more permissive environment for survival and integration of transplanted cells. In this study, fibrin hydrogels functionalized with a synthetic peptide engaging integrin α6β1 (HYD1) were shown to promote neurite extension of ES-NSPCs, which is fundamental for the formation of functional neuronal relay circuits after NSPC transplantation. Notably, HYD1-functionalized fibrin per se led to enhanced axonal growth ex vivo and to an improvement in locomotor function after implantation in a rat model of spinal cord injury. Conjugation of α6β1 integrin-binding motifs may therefore be of interest to confer bioactivity to NSPC hydrogel vehicles.
Collapse
Affiliation(s)
- Joana Silva
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Ana R Bento
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Portugal
| | - Daniela Barros
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Tiago L Laundos
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Susana R Sousa
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; ISEP - Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Portugal
| | - Pedro Quelhas
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Mónica M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Nerve Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
| | - Ana P Pêgo
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Isabel F Amaral
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Portugal.
| |
Collapse
|
31
|
Nawrotek K, Marqueste T, Modrzejewska Z, Zarzycki R, Rusak A, Decherchi P. Thermogelling chitosan lactate hydrogel improves functional recovery after a C2 spinal cord hemisection in rat. J Biomed Mater Res A 2017; 105:2004-2019. [PMID: 28324618 DOI: 10.1002/jbm.a.36067] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/23/2017] [Accepted: 03/15/2017] [Indexed: 11/06/2022]
Abstract
The present study was designed to provide an appropriate micro-environment for regenerating axotomized neurons and proliferating/migrating cells. Because of its intrinsic permissive properties, biocompatibility and biodegradability, we chose to evaluate the therapeutic effectiveness of a chitosan-based biopolymer. The biomaterial toxicity was measured through in vitro test based on fibroblast cell survival on thermogelling chitosan lactate hydrogel substrate and then polymer was implanted into a C2 hemisection of the rat spinal cord. Animals were randomized into three experimental groups (Control, Lesion and Lesion + Hydrogel) and functional tests (ladder walking and forelimb grip strength tests, respiratory assessment by whole-body plethysmography measurements) were used, once a week during 10 weeks, to evaluate post-traumatic recoveries. Then, electrophysiological examinations (reflexivity of the sub-lesional region, ventilatory adjustments to muscle fatigue known to elicit the muscle metaboreflex and phrenic nerve recordings during normoxia and temporary hypoxia) were performed. In vitro results indicated that the chitosan matrix is a non-toxic biomaterial that allowed fibroblast survival. Furthermore, implanted animals showed improvements of their ladder walking scores from the 4th week post-implantation. Finally, electrophysiological recordings indicated that animals receiving the chitosan matrix exhibited recovery of the H-reflex rate sensitive depression, the ventilatory response to repetitive muscle stimulation and an increase of the phrenic nerve activity to asphyxia compared to lesioned and nonimplanted animals. This study indicates that hydrogel based on chitosan constitute a promising therapeutic approach to repair damaged spinal cord or may be used as an adjuvant with other treatments to enhance functional recovery after a central nervous system damage. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2004-2019, 2017.
Collapse
Affiliation(s)
- Katarzyna Nawrotek
- Faculty of Process and Environmental Engineering, Department of Chemical Engineering, Lodz University of Technology, Wolczanska 175 Street, Lodz, 90-924, Poland
| | - Tanguy Marqueste
- Aix-Marseille Université (AMU) and Centre National de la Recherche Scientifique (CNRS), Institut des Sciences du Mouvement (UMR 7287), Equipe « Plasticité des Systèmes Nerveux et Musculaire », Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288, Marseille Cedex 09, France
| | - Zofia Modrzejewska
- Faculty of Process and Environmental Engineering, Department of Chemical Engineering, Lodz University of Technology, Wolczanska 175 Street, Lodz, 90-924, Poland
| | - Roman Zarzycki
- Faculty of Process and Environmental Engineering, Department of Chemical Engineering, Lodz University of Technology, Wolczanska 175 Street, Lodz, 90-924, Poland
| | - Agnieszka Rusak
- Department of Experimental Surgery and Biomaterials Research, Wroclaw Medical University, Medico-Dental Faculty, Krakowska 26 Street, Wroclaw, Poland, 50-425
| | - Patrick Decherchi
- Aix-Marseille Université (AMU) and Centre National de la Recherche Scientifique (CNRS), Institut des Sciences du Mouvement (UMR 7287), Equipe « Plasticité des Systèmes Nerveux et Musculaire », Parc Scientifique et Technologique de Luminy, CC910-163, Avenue de Luminy, F-13288, Marseille Cedex 09, France
| |
Collapse
|
32
|
Ordikhani F, Sheth S, Zustiak SP. Polymeric particle-mediated molecular therapies to treat spinal cord injury. Int J Pharm 2017; 516:71-81. [DOI: 10.1016/j.ijpharm.2016.11.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 11/03/2016] [Accepted: 11/08/2016] [Indexed: 11/26/2022]
|
33
|
Bento AR, Quelhas P, Oliveira MJ, Pêgo AP, Amaral IF. Three-dimensional culture of single embryonic stem-derived neural/stem progenitor cells in fibrin hydrogels: neuronal network formation and matrix remodelling. J Tissue Eng Regen Med 2016; 11:3494-3507. [PMID: 28032468 DOI: 10.1002/term.2262] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 06/15/2016] [Accepted: 07/03/2016] [Indexed: 12/20/2022]
Abstract
In an attempt to improve the efficacy of neural stem/progenitor cell (NSPC) based therapies, fibrin hydrogels are being explored to provide a favourable microenvironment for cell survival and differentiation following transplantation. In the present work, the ability of fibrin to support the survival, proliferation, and neuronal differentiation of NSPCs derived from embryonic stem (ES) cells under monolayer culture was explored. Single mouse ES-NSPCs were cultured within fibrin (fibrinogen concentration: 6 mg/ml) under neuronal differentiation conditions up to 14 days. The ES-NSPCs retained high cell viability and proliferated within small-sized spheroids. Neuronal differentiation was confirmed by an increase in the levels of βIII-tubulin and NF200 over time. At day 14, cell-matrix constructs mainly comprised NSPCs and neurons (46.5% βIII-tubulin+ cells). Gamma-aminobutyric acid (GABA)ergic and dopaminergic/noradrenergic neurons were also observed, along with a network of synaptic proteins. The ES-NSPCs expressed matriptase and secreted MMP-2/9, with MMP-2 activity increasing along time. Fibronectin, laminin and collagen type IV deposition was also detected. Fibrin gels prepared with higher fibrinogen concentrations (8/10 mg/ml) were less permissive to neurite extension and neuronal differentiation, possibly owing to their smaller pore area and higher rigidity. Overall, it is shown that ES-NSPCs within fibrin are able to establish neuronal networks and to remodel fibrin through MMP secretion and extracellular matrix (ECM) deposition. This three-dimensional (3D) culture system was also shown to support cell viability, neuronal differentiation and ECM deposition of human ES-NSPCs. The settled 3D platform is expected to constitute a valuable tool to develop fibrin-based hydrogels for ES-NSPC delivery into the injured central nervous system. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ana R Bento
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Faculdade de Engenharia, Universidade do Porto, Portugal
| | - Pedro Quelhas
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Maria J Oliveira
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Departamento de Patologia e Oncologia da Faculdade de Medicina da Universidade do Porto de Medicina da Universidade do Porto, Portugal
| | - Ana P Pêgo
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Faculdade de Engenharia, Universidade do Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Isabel F Amaral
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal.,Faculdade de Engenharia, Universidade do Porto, Portugal
| |
Collapse
|
34
|
Tukmachev D, Forostyak S, Koci Z, Zaviskova K, Vackova I, Vyborny K, Sandvig I, Sandvig A, Medberry CJ, Badylak SF, Sykova E, Kubinova S. Injectable Extracellular Matrix Hydrogels as Scaffolds for Spinal Cord Injury Repair. Tissue Eng Part A 2016; 22:306-17. [PMID: 26729284 DOI: 10.1089/ten.tea.2015.0422] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Restoration of lost neuronal function after spinal cord injury (SCI) still remains a big challenge for current medicine. One important repair strategy is bridging the SCI lesion with a supportive and stimulatory milieu that would enable axonal rewiring. Injectable extracellular matrix (ECM)-derived hydrogels have been recently reported to have neurotrophic potential in vitro. In this study, we evaluated the presumed neuroregenerative properties of ECM hydrogels in vivo in the acute model of SCI. ECM hydrogels were prepared by decellularization of porcine spinal cord (SC) or porcine urinary bladder (UB), and injected into a spinal cord hemisection cavity. Histological analysis and real-time qPCR were performed at 2, 4, and 8 weeks postinjection. Both types of hydrogels integrated into the lesion and stimulated neovascularization and axonal ingrowth into the lesion. On the other hand, massive infiltration of macrophages into the lesion and rapid hydrogel degradation did not prevent cyst formation, which progressively developed over 8 weeks. No significant differences were found between SC-ECM and UB-ECM. Gene expression analysis revealed significant downregulation of genes related to immune response and inflammation in both hydrogel types at 2 weeks post SCI. A combination of human mesenchymal stem cells with SC-ECM did not further promote ingrowth of axons and blood vessels into the lesion, when compared with the SC-ECM hydrogel alone. In conclusion, both ECM hydrogels bridged the lesion cavity, modulated the innate immune response, and provided the benefit of a stimulatory substrate for in vivo neural tissue regeneration. However, fast hydrogel degradation might be a limiting factor for the use of native ECM hydrogels in the treatment of acute SCI.
Collapse
Affiliation(s)
- Dmitry Tukmachev
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Serhiy Forostyak
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Zuzana Koci
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Kristyna Zaviskova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Irena Vackova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic
| | - Karel Vyborny
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Ioanna Sandvig
- 3 Department of Neuroscience, Norwegian University of Science and Technology , Trondheim, Norway .,4 John Van Geest Centre for Brain Repair, School of Clinical Neurosciences, University of Cambridge , Cambridge, United Kingdom
| | - Axel Sandvig
- 3 Department of Neuroscience, Norwegian University of Science and Technology , Trondheim, Norway .,5 Division of Pharmacology and Clinical Neuroscience, Department of Neurosurgery, Umeå University , Umeå, Sweden
| | | | - Stephen F Badylak
- 6 McGowan Institute for Regenerative Medicine , Pittsburgh, Pennsylvania
| | - Eva Sykova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic .,2 2nd Medical Faculty, Charles University , Prague, Czech Republic
| | - Sarka Kubinova
- 1 Institute of Experimental Medicine AS CR , Prague, Czech Republic
| |
Collapse
|
35
|
Breen BA, Kraskiewicz H, Ronan R, Kshiragar A, Patar A, Sargeant T, Pandit A, McMahon SS. Therapeutic Effect of Neurotrophin-3 Treatment in an Injectable Collagen Scaffold Following Rat Spinal Cord Hemisection Injury. ACS Biomater Sci Eng 2016; 3:1287-1295. [DOI: 10.1021/acsbiomaterials.6b00167] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | - Timothy Sargeant
- Covidien LLC, 60 Middletown Avenue, North Haven, Connecticut 06473, United States
| | | | | |
Collapse
|
36
|
Arai K, Harada Y, Tomiyama H, Michishita M, Kanno N, Yogo T, Suzuki Y, Hara Y. Evaluation of the survival of bone marrow-derived mononuclear cells and the growth factors produced upon intramedullary transplantation in rat models of acute spinal cord injury. Res Vet Sci 2016; 107:88-94. [PMID: 27473980 DOI: 10.1016/j.rvsc.2016.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 05/19/2016] [Accepted: 05/22/2016] [Indexed: 01/13/2023]
Abstract
Intramedullary bone marrow-derived mononuclear cell (BM-MNC) transplantation has demonstrated neuroprotective effects in the chronic stage of spinal cord injury (SCI). However, no previous study has evaluated its effects in the acute stage, even though cell death occurs mainly within 1week after injury in all neuronal cells. Moreover, the mechanism underlying these effects remains unclear. We aimed to investigate the survival of intramedullary transplanted allogeneic BM-MNCs and the production of growth factors after transplantation to clarify the therapeutic potential of intramedullary transplanted BM-MNCs and their protective effects in acute SCI. Sprague-Dawley rats were subjected to traumatic SCI and received intramedullary transplantation of EGFP(+)BM-MNCs (n=6), BM-MNCs (n=10), or solvent (n=10) immediately after injury. To evaluate the transplanted BM-MNCs and their therapeutic effects, immunohistochemical evaluations were performed at 3 and 7days post-injury (DPI). BM-MNCs were observed at the injected site at both 3 (683±83 cells/mm(2)) and 7 DPI (395±64 cells/mm(2)). The expression of hepatocyte growth factor was observed in approximately 20% transplanted BM-MNCs. Some BM-MNCs also expressed monocyte chemotactic protein-1 or vascular endothelial growth factor. The demyelinated area and number of cleaved caspase-3-positive cells were significantly smaller in the BM-MNC-transplanted group at 3 DPI. Hindlimb locomotor function was significantly improved in the BM-MNC-transplanted group at 7 DPI. These results suggest that intramedullary transplantation of BM-MNCs is an efficient method for introducing a large number of growth factor-producing cells that can induce neuroprotective effects in the acute stage of SCI.
Collapse
Affiliation(s)
- Kiyotaka Arai
- Laboratory of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Yasuji Harada
- Laboratory of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan.
| | - Hiroyuki Tomiyama
- Laboratory of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Masaki Michishita
- Laboratory of Veterinary Pathology, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Nobuo Kanno
- Laboratory of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Takuya Yogo
- Laboratory of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| | - Yoshihisa Suzuki
- Department of Plastic and Reconstructive Surgery, Kitano Hospital, Tazuke Kofukai Medical Research Institute, Osaka 530-8507, Japan
| | - Yasushi Hara
- Laboratory of Veterinary Surgery, Nippon Veterinary and Life Science University, 1-7-1, Kyounan-cho, Musashino, Tokyo 180-8602, Japan
| |
Collapse
|
37
|
Ge L, Liu K, Liu Z, Lu M. Co-transplantation of autologous OM-MSCs and OM-OECs: a novel approach for spinal cord injury. Rev Neurosci 2016; 27:259-70. [PMID: 26574889 DOI: 10.1515/revneuro-2015-0030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/09/2015] [Indexed: 11/15/2022]
Abstract
AbstractSpinal cord injury (SCI) is a disastrous injury that leads to motor and sensory dysfunctions in patients. In recent years, co-transplantation has become an increasingly used therapeutic treatment for patients with SCI. Both mesenchymal stem cells (MSCs) and olfactory-ensheathing cells (OECs) have been adopted to ameliorate SCI, with promising outcomes. Remarkable effects on the rehabilitation of patients with SCI have been achieved using MSCs. Olfactory mucosa (OM) MSCs from human OM are one of the most ideal cell resources for auto-transplantation in clinical application owing to their a high proliferation rate and multipotent capability. In addition, OECs derived from OM have been used to improve functional recovery of SCI and resulted in promising functional recovery in years. Accordingly, co-transplantation of OM-MSCs coupled with OM-OECs has been adopted to improve the recovery of SCI. Here we reviewed the reported applications of OM-MSCs and OM-OECs for SCI treatment and proposed that a novel combined strategy using both autologous OM-MSCs and OM-OECs would achieve a better approach for the treatment of SCI.
Collapse
Affiliation(s)
| | | | - Zhonghua Liu
- 2College of Life Sciences, Hunan Normal University, Changsha 410008, P.R. China
| | - Ming Lu
- 1Department of Neurosurgery, Second Affiliated Hospital of Hunan Normal University (163 Hospital of PLA), Changsha 410003, P.R. China
| |
Collapse
|
38
|
Desautels TA, Choe J, Gad P, Nandra MS, Roy RR, Zhong H, Tai YC, Edgerton VR, Burdick JW. An Active Learning Algorithm for Control of Epidural Electrostimulation. IEEE Trans Biomed Eng 2015; 62:2443-2455. [PMID: 25974925 PMCID: PMC4617183 DOI: 10.1109/tbme.2015.2431911] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Epidural electrostimulation has shown promise for spinal cord injury therapy. However, finding effective stimuli on the multi-electrode stimulating arrays employed requires a laborious manual search of a vast space for each patient. Widespread clinical application of these techniques would be greatly facilitated by an autonomous, algorithmic system which choses stimuli to simultaneously deliver effective therapy and explore this space. We propose a method based on GP-BUCB, a Gaussian process bandit algorithm. In n = 4 spinally transected rats, we implant epidural electrode arrays and examine the algorithm's performance in selecting bipolar stimuli to elicit specified muscle responses. These responses are compared with temporally interleaved intra-animal stimulus selections by a human expert. GP-BUCB successfully controlled the spinal electrostimulation preparation in 37 testing sessions, selecting 670 stimuli. These sessions included sustained autonomous operations (ten-session duration). Delivered performance with respect to the specified metric was as good as or better than that of the human expert. Despite receiving no information as to anatomically likely locations of effective stimuli, GP-BUCB also consistently discovered such a pattern. Further, GP-BUCB was able to extrapolate from previous sessions' results to make predictions about performance in new testing sessions, while remaining sufficiently flexible to capture temporal variability. These results provide validation for applying automated stimulus selection methods to the problem of spinal cord injury therapy.
Collapse
Affiliation(s)
- Thomas A Desautels
- California Institute of Technology, Pasadena, CA 91125, USA. He is now with the Gatsby Computational Neuroscience Unit, University College London, London WC1N 3AR UK
| | - Jaehoon Choe
- University of California, Los Angeles, Los Angeles, CA, USA. J. Choe is now with HRL Laboratories, LLC, Malibu, CA, USA
| | - Parag Gad
- University of California, Los Angeles, Los Angeles, CA, USA. J. Choe is now with HRL Laboratories, LLC, Malibu, CA, USA
| | | | - Roland R Roy
- University of California, Los Angeles, Los Angeles, CA, USA. J. Choe is now with HRL Laboratories, LLC, Malibu, CA, USA
| | - Hui Zhong
- University of California, Los Angeles, Los Angeles, CA, USA. J. Choe is now with HRL Laboratories, LLC, Malibu, CA, USA
| | - Yu-Chong Tai
- California Institute of Technology, Pasadena, CA, USA
| | - V Reggie Edgerton
- University of California, Los Angeles, Los Angeles, CA, USA. J. Choe is now with HRL Laboratories, LLC, Malibu, CA, USA
| | | |
Collapse
|
39
|
Raspa A, Pugliese R, Maleki M, Gelain F. Recent therapeutic approaches for spinal cord injury. Biotechnol Bioeng 2015; 113:253-9. [DOI: 10.1002/bit.25689] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 06/21/2015] [Accepted: 06/24/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Andrea Raspa
- Center for Nanomedicine and Tissue Engineering (CNTE); A. O. Ospedale Niguarda Cà Granda, Piazza dell' ospedale maggiore 3, 20162 Milan; Italy
- IRCCS Casa Sollievo della Sofferenza, Opera di San Pio da Pietralcina; Viale Capuccini 1 San Giovanni Rotondo (FG) 71013; Italy
| | - Raffaele Pugliese
- Center for Nanomedicine and Tissue Engineering (CNTE); A. O. Ospedale Niguarda Cà Granda, Piazza dell' ospedale maggiore 3, 20162 Milan; Italy
- IRCCS Casa Sollievo della Sofferenza, Opera di San Pio da Pietralcina; Viale Capuccini 1 San Giovanni Rotondo (FG) 71013; Italy
| | - Mahboubeh Maleki
- Center for Nanomedicine and Tissue Engineering (CNTE); A. O. Ospedale Niguarda Cà Granda, Piazza dell' ospedale maggiore 3, 20162 Milan; Italy
- IRCCS Casa Sollievo della Sofferenza, Opera di San Pio da Pietralcina; Viale Capuccini 1 San Giovanni Rotondo (FG) 71013; Italy
| | - Fabrizio Gelain
- Center for Nanomedicine and Tissue Engineering (CNTE); A. O. Ospedale Niguarda Cà Granda, Piazza dell' ospedale maggiore 3, 20162 Milan; Italy
- IRCCS Casa Sollievo della Sofferenza, Opera di San Pio da Pietralcina; Viale Capuccini 1 San Giovanni Rotondo (FG) 71013; Italy
| |
Collapse
|
40
|
Pires LR, Pêgo AP. Bridging the lesion-engineering a permissive substrate for nerve regeneration. Regen Biomater 2015; 2:203-14. [PMID: 26816642 PMCID: PMC4669012 DOI: 10.1093/rb/rbv012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/21/2015] [Accepted: 06/30/2015] [Indexed: 01/30/2023] Open
Abstract
Biomaterial-based strategies to restore connectivity after lesion at the spinal cord are focused on bridging the lesion and providing an favourable substrate and a path for axonal re-growth. Following spinal cord injury (SCI) a hostile environment for neuronal cell growth is established by the activation of multiple inhibitory mechanisms that hamper regeneration to occur. Implantable scaffolds can provide mechanical support and physical guidance for axon re-growth and, at the same time, contribute to alleviate the hostile environment by the in situ delivery of therapeutic molecules and/or relevant cells. Basic research on SCI has been contributing with the description of inhibitory mechanisms for regeneration as well as identifying drugs/molecules that can target inhibition. This knowledge is the background for the development of combined strategies with biomaterials. Additionally, scaffold design is significantly evolving. From the early simple hollow conduits, scaffolds with complex architectures that can modulate cell fate are currently being tested. A number of promising pre-clinical studies combining scaffolds, cells, drugs and/or nucleic acids are reported in the open literature. Overall, it is considered that to address the multi-factorial inhibitory environment of a SCI, a multifaceted therapeutic approach is imperative. The progress in the identification of molecules that target inhibition after SCI and its combination with scaffolds and/or cells are described and discussed in this review.
Collapse
Affiliation(s)
- Liliana R. Pires
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Faculdade de Engenharia—Universidade do Porto (FEUP), Porto, Portugal and
| | - Ana P. Pêgo
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Faculdade de Engenharia—Universidade do Porto (FEUP), Porto, Portugal and
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
41
|
Buzhor E, Leshansky L, Blumenthal J, Barash H, Warshawsky D, Mazor Y, Shtrichman R. Cell-based therapy approaches: the hope for incurable diseases. Regen Med 2015; 9:649-72. [PMID: 25372080 DOI: 10.2217/rme.14.35] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cell therapies aim to repair the mechanisms underlying disease initiation and progression, achieved through trophic effect or by cell replacement. Multiple cell types can be utilized in such therapies, including stem, progenitor or primary cells. This review covers the current state of cell therapies designed for the prominent disorders, including cardiovascular, neurological (Parkinson's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury), autoimmune (Type 1 diabetes, multiple sclerosis, Crohn's disease), ophthalmologic, renal, liver and skeletal (osteoarthritis) diseases. Various cell therapies have reached advanced clinical trial phases with potential marketing approvals in the near future, many of which are based on mesenchymal stem cells. Advances in pluripotent stem cell research hold great promise for regenerative medicine. The information presented in this review is based on the analysis of the cell therapy collection detailed in LifeMap Discovery(®) (LifeMap Sciences Inc., USA) the database of embryonic development, stem cell research and regenerative medicine.
Collapse
|
42
|
Hydrogels and Cell Based Therapies in Spinal Cord Injury Regeneration. Stem Cells Int 2015; 2015:948040. [PMID: 26124844 PMCID: PMC4466497 DOI: 10.1155/2015/948040] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/14/2014] [Indexed: 01/01/2023] Open
Abstract
Spinal cord injury (SCI) is a central nervous system- (CNS-) related disorder for which there is yet no successful treatment. Within the past several years, cell-based therapies have been explored for SCI repair, including the use of pluripotent human stem cells, and a number of adult-derived stem and mature cells such as mesenchymal stem cells, olfactory ensheathing cells, and Schwann cells. Although promising, cell transplantation is often overturned by the poor cell survival in the treatment of spinal cord injuries. Alternatively, the therapeutic role of different cells has been used in tissue engineering approaches by engrafting cells with biomaterials. The latter have the advantages of physically mimicking the CNS tissue, while promoting a more permissive environment for cell survival, growth, and differentiation. The roles of both cell- and biomaterial-based therapies as single therapeutic approaches for SCI repair will be discussed in this review. Moreover, as the multifactorial inhibitory environment of a SCI suggests that combinatorial approaches would be more effective, the importance of using biomaterials as cell carriers will be herein highlighted, as well as the recent advances and achievements of these promising tools for neural tissue regeneration.
Collapse
|
43
|
van der Kallen LR, Eggers R, Ehlert EM, Verhaagen J, Smit AB, van Kesteren RE. Genetic Deletion of the Transcriptional Repressor NFIL3 Enhances Axon Growth In Vitro but Not Axonal Repair In Vivo. PLoS One 2015; 10:e0127163. [PMID: 25993115 PMCID: PMC4438979 DOI: 10.1371/journal.pone.0127163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/13/2015] [Indexed: 01/04/2023] Open
Abstract
Axonal regeneration after injury requires the coordinated expression of genes in injured neurons. We previously showed that either reducing expression or blocking function of the transcriptional repressor NFIL3 activates transcription of regeneration-associated genes Arg1 and Gap43 and strongly promotes axon outgrowth in vitro. Here we tested whether genetic deletion or dominant-negative inhibition of NFIL3 could promote axon regeneration and functional recovery after peripheral nerve lesion in vivo. Contrary to our expectations, we observed no changes in the expression of regeneration-associated genes and a significant delay in functional recovery following genetic deletion of Nfil3. When NFIL3 function was inhibited specifically in dorsal root ganglia prior to sciatic nerve injury, we observed a decrease in regenerative axon growth into the distal nerve segment rather than an increase. Finally, we show that deletion of Nfil3 changes sciatic nerve lesion-induced expression in dorsal root ganglia of genes that are not typically involved in regeneration, including several olfactory receptors and developmental transcription factors. Together our findings show that removal of NFIL3 in vivo does not recapitulate the regeneration-promoting effects that were previously observed in vitro, indicating that in vivo transcriptional control of regeneration is probably more complex and more robust against perturbation than in vitro data may suggest.
Collapse
Affiliation(s)
- Loek R. van der Kallen
- Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ruben Eggers
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Erich M. Ehlert
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - August B. Smit
- Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ronald E. van Kesteren
- Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
44
|
Tukmachev D, Lunov O, Zablotskii V, Dejneka A, Babic M, Syková E, Kubinová Š. An effective strategy of magnetic stem cell delivery for spinal cord injury therapy. NANOSCALE 2015; 7:3954-8. [PMID: 25652717 DOI: 10.1039/c4nr05791k] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Spinal cord injury (SCI) is a condition that results in significant mortality and morbidity. Treatment of SCI utilizing stem cell transplantation represents a promising therapy. However, current conventional treatments are limited by inefficient delivery strategies of cells into the injured tissue. In this study, we designed a magnetic system and used it to accumulate stem cells labelled with superparamagnetic iron oxide nanoparticles (SPION) at a specific site of a SCI lesion. The loading of stem cells with engineered SPIONs that guarantees sufficient attractive magnetic forces was achieved. Further, the magnetic system allowed rapid guidance of the SPION-labelled cells precisely to the lesion location. Histological analysis of cell distribution throughout the cerebrospinal channel showed a good correlation with the calculated distribution of magnetic forces exerted onto the transplanted cells. The results suggest that focused targeting and fast delivery of stem cells can be achieved using the proposed non-invasive magnetic system. With future implementation the proposed targeting and delivery strategy bears advantages for the treatment of disease requiring fast stem cell transplantation.
Collapse
Affiliation(s)
- Dmitry Tukmachev
- Institute of Experimental Medicine, ASCR, 14200 Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
ABSTRACT Restoration of lost neuronal function after spinal cord injury still remains a considerable challenge for current medicine. Over the last decade, regenerative medicine has recorded rapid and promising advancements in stem cell research, genetic engineering and the progression of new sophisticated biomaterials as well as nanotechnology. This advancement has also been reflected in neural tissue engineering, where, along with the development of a new generation of well-designed biopolymer scaffolds, multifactorial therapeutic strategies are being validated in order to determine the greatest possible repair efficacy of the complex CNS pathophysiology. Much attention is currently focused on the designing of multifunctional polymer scaffolds as systems for targeted drug or gene delivery, electrical stimulation or as substrates creating a special micro-environment, promoting the growth and desired differentiation of various cell lines. In this review, the latest advances in biomaterial technology together with various combinatorial strategies designed to treat spinal cord injury treatment are summarized and discussed.
Collapse
|
46
|
Astrocytes increase ATP exocytosis mediated calcium signaling in response to microgroove structures. Sci Rep 2015; 5:7847. [PMID: 25597401 PMCID: PMC4297955 DOI: 10.1038/srep07847] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/16/2014] [Indexed: 01/14/2023] Open
Abstract
Following central nervous system (CNS) injury, activated astrocytes form glial scars, which inhibit axonal regeneration, leading to long-term functional deficits. Engineered nanoscale scaffolds guide cell growth and enhance regeneration within models of spinal cord injury. However, the effects of micro-/nanosize scaffolds on astrocyte function are not well characterized. In this study, a high throughput (HTP) microscale platform was developed to study astrocyte cell behavior on micropatterned surfaces containing 1 μm spacing grooves with a depth of 250 or 500 nm. Significant changes in cell and nuclear elongation and alignment on patterned surfaces were observed, compared to on flat surfaces. The cytoskeleton components (particularly actin filaments and focal adhesions) and nucleus-centrosome axis were aligned along the grooved direction as well. More interestingly, astrocytes on micropatterned surfaces showed enhanced mitochondrial activity with lysosomes localized at the lamellipodia of the cells, accompanied by enhanced adenosine triphosphate (ATP) release and calcium activities. These data indicate that the lysosome-mediated ATP exocytosis and calcium signaling may play an important role in astrocytic responses to substrate topology. These new findings have furthered our understanding of the biomechanical regulation of astrocyte cell–substrate interactions, and may benefit the optimization of scaffold design for CNS healing.
Collapse
|
47
|
Li Y, Alam M, Guo S, Ting KH, He J. Electronic bypass of spinal lesions: activation of lower motor neurons directly driven by cortical neural signals. J Neuroeng Rehabil 2014; 11:107. [PMID: 24990580 PMCID: PMC4094416 DOI: 10.1186/1743-0003-11-107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 06/20/2014] [Indexed: 01/08/2023] Open
Abstract
Background Lower motor neurons in the spinal cord lose supraspinal inputs after complete spinal cord injury, leading to a loss of volitional control below the injury site. Extensive locomotor training with spinal cord stimulation can restore locomotion function after spinal cord injury in humans and animals. However, this locomotion is non-voluntary, meaning that subjects cannot control stimulation via their natural “intent”. A recent study demonstrated an advanced system that triggers a stimulator using forelimb stepping electromyographic patterns to restore quadrupedal walking in rats with spinal cord transection. However, this indirect source of “intent” may mean that other non-stepping forelimb activities may false-trigger the spinal stimulator and thus produce unwanted hindlimb movements. Methods We hypothesized that there are distinguishable neural activities in the primary motor cortex during treadmill walking, even after low-thoracic spinal transection in adult guinea pigs. We developed an electronic spinal bridge, called “Motolink”, which detects these neural patterns and triggers a “spinal” stimulator for hindlimb movement. This hardware can be head-mounted or carried in a backpack. Neural data were processed in real-time and transmitted to a computer for analysis by an embedded processor. Off-line neural spike analysis was conducted to calculate and preset the spike threshold for “Motolink” hardware. Results We identified correlated activities of primary motor cortex neurons during treadmill walking of guinea pigs with spinal cord transection. These neural activities were used to predict the kinematic states of the animals. The appropriate selection of spike threshold value enabled the “Motolink” system to detect the neural “intent” of walking, which triggered electrical stimulation of the spinal cord and induced stepping-like hindlimb movements. Conclusion We present a direct cortical “intent”-driven electronic spinal bridge to restore hindlimb locomotion after complete spinal cord injury.
Collapse
Affiliation(s)
| | | | | | | | - Jufang He
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.
| |
Collapse
|
48
|
An in vitro spinal cord injury model to screen neuroregenerative materials. Biomaterials 2014; 35:3756-65. [DOI: 10.1016/j.biomaterials.2014.01.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/08/2014] [Indexed: 02/06/2023]
|
49
|
Rocha DN, Brites P, Fonseca C, Pêgo AP. Poly(trimethylene carbonate-co-ε-caprolactone) promotes axonal growth. PLoS One 2014; 9:e88593. [PMID: 24586346 PMCID: PMC3937290 DOI: 10.1371/journal.pone.0088593] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 01/13/2014] [Indexed: 12/31/2022] Open
Abstract
Mammalian central nervous system (CNS) neurons do not regenerate after injury due to the inhibitory environment formed by the glial scar, largely constituted by myelin debris. The use of biomaterials to bridge the lesion area and the creation of an environment favoring axonal regeneration is an appealing approach, currently under investigation. This work aimed at assessing the suitability of three candidate polymers – poly(ε-caprolactone), poly(trimethylene carbonate-co-ε-caprolactone) (P(TMC-CL)) (11∶89 mol%) and poly(trimethylene carbonate) - with the final goal of using these materials in the development of conduits to promote spinal cord regeneration. Poly(L-lysine) (PLL) coated polymeric films were tested for neuronal cell adhesion and neurite outgrowth. At similar PLL film area coverage conditions, neuronal polarization and axonal elongation was significantly higher on P(TMC-CL) films. Furthermore, cortical neurons cultured on P(TMC-CL) were able to extend neurites even when seeded onto myelin. This effect was found to be mediated by the glycogen synthase kinase 3β (GSK3β) signaling pathway with impact on the collapsin response mediator protein 4 (CRMP4), suggesting that besides surface topography, nanomechanical properties were implicated in this process. The obtained results indicate P(TMC-CL) as a promising material for CNS regenerative applications as it promotes axonal growth, overcoming myelin inhibition.
Collapse
Affiliation(s)
- Daniela Nogueira Rocha
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Pedro Brites
- Nerve Regeneration Group, IBMC – Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Carlos Fonseca
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
50
|
Electroacupuncture improves bladder and bowel function in patients with traumatic spinal cord injury: results from a prospective observational study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:543174. [PMID: 24382977 PMCID: PMC3870613 DOI: 10.1155/2013/543174] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Accepted: 11/14/2013] [Indexed: 11/22/2022]
Abstract
In order to explore the effect of electroacupuncture (EA) for chronic bowel and bladder dysfunction after traumatic spinal cord injury, 14 patients were treated with electroacupuncture once a day, five times a week for the first four weeks, and once every other day, three times a week for the following four weeks. The patients were then followed up for six months. After treatment, four (4/14, 28.57%) patients resumed normal voiding; six (6/14, 42.86%) resumed normal voiding for no less than half of all micturition behaviors; four (4/14, 28.57%) required supplementary urination methods for higher than half of all micturition behaviors. These effects persisted during followup. Mean postvoid RUV decreased by 190.29 ± 101.87 mL (P < 0.01) after treatment and by 198.86 ± 112.18 mL (P < 0.01) during followup. Patients' weekly urinary incontinence frequency decreased 7.14 ± 46.34 times/week (P = 0.036) after treatment and decreased 49.86 ± 44.38 times/week during followup. After treatment, four (4/14, 28.57%) patients resumed normal bowel movements (P = 0.025); five (5/14, 35.71%) reduced the dependence on supplementary defecation methods; five (5/14, 35.71%) had no changes. In patients with chronic bowel and bladder dysfunction after traumatic SCI, EA may provide a valuable alternative tool in improving patients' self-controlled bowel and bladder functions with minimal side effects.
Collapse
|