1
|
Jung JW, Jeong JH, Ko MJ, Lee BJ, Kwon WK, Jeon SR, Lee S. Induced Neural Stem Cell Transplantation in Spinal Cord Injury: Present Status and Next Steps. Korean J Neurotrauma 2024; 20:234-245. [PMID: 39803345 PMCID: PMC11711022 DOI: 10.13004/kjnt.2024.20.e45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Spinal cord injury (SCI) remains a significant clinical challenge, with no fully effective treatment available despite advancements in various therapeutic approaches. This review examines the emerging role of induced neural stem cells (iNSCs) as promising candidates for SCI treatment, highlighting their potential for direct neural regeneration and integration with host tissue. We explore the biology of iNSCs, their mechanisms of action, and their interactions with host tissue, including modulating inflammatory responses, promoting axonal growth, and reconstructing neural circuits. Additionally, the importance of administration route, optimal timing for transplantation, and potential adverse events are discussed to address key challenges in translating these therapies to clinical applications. The review also emphasizes recent innovations, such as combining iNSC transplantation with rehabilitative training and the integration of biomaterials and growth factors to enhance therapeutic efficacy. Although preclinical studies have demonstrated positive outcomes, larger, controlled trials and standardized protocols are essential for validating the safety and effectiveness of iNSC-based therapies for SCI patients.
Collapse
Affiliation(s)
- Jae-Woo Jung
- Department of Neurosurgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Je Hoon Jeong
- Department of Neurosurgery, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Myeong Jin Ko
- Department of Neurosurgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Korea
| | - Byung-Jou Lee
- Department of Neurosurgery, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Woo-Keun Kwon
- Department of Neurosurgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Sang Ryong Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Subum Lee
- Department of Neurosurgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Ait Hamdan Y, El-Mansoury B, Elouali S, Rachmoune K, Belbachir A, Oudadesse H, Rhazi M. A review of chitosan polysaccharides: Neuropharmacological implications and tissue regeneration. Int J Biol Macromol 2024; 279:135356. [PMID: 39244136 DOI: 10.1016/j.ijbiomac.2024.135356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
One of the current challenges in targeting neurological disorders is that many therapeutic molecules cannot cross the blood-brain barrier (BBB), which limits the use of natural molecules in nervous tissue regeneration. Thus, the development of new drugs to effectively treat neurological disorders would be a challenge. Natural resources are well known as a source of several therapeutic agents for the treatment of neurologic disorders. Recently, chitosan (CTS) and its derivatives from arthropod exoskeletons, have attracted much attention as a drug delivery system to transport therapeutic substances across the BBB and thanks to other neuroprotective effects including the participation to the CNS regenerations scaffolds to replicate the extracellular matrix and microenvironment of the body. This review will discuss the place of natural resource therapy in targeting neurological disorders. In particular, it will highlight recent understanding and progress in the applications of CTS as drug delivery systems and their therapeutic effects on these disorders through tissue regeneration, as well as the molecular mechanisms by which they exert these effects.
Collapse
Affiliation(s)
- Youssef Ait Hamdan
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; Univ Rennes, CNRS, ISCR-UMR 6226, F-35000 Rennes, France.
| | - Bilal El-Mansoury
- Laboratory of Anthropogenic, Biotechnology and Health, Team physiopathology Nutritional, Neurosciences and Toxicology, Faculty of Sciences, Chouaib Doukkali University, Av. Des facultés, 24000 El Jadida, Morocco
| | - Samia Elouali
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; University of Mons (UMONS) - Laboratory of Polymeric and Composite Materials (LPCM), Center of Innovation and Research in Materials and Polymers (CIRMAP), Place du Parc 20, 7000 Mons, Belgium
| | - Khawla Rachmoune
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco; Biotechnology and Biomolecule Engineering Unit, CNESTEN, Rabat, Morocco
| | - Anass Belbachir
- Center for Regenerative Medicine, CHU MOHAMMED VI, Marrakech, Morocco; Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | | | - Mohammed Rhazi
- Interdisciplinary Laboratory of Research in Bio-Resources, Environment and Materials, Higher Normal School, Cadi Ayyad University, 40000 Marrakech, Morocco
| |
Collapse
|
3
|
Ralph PC, Choi SW, Baek MJ, Lee SJ. Regenerative medicine approaches for the treatment of spinal cord injuries: Progress and challenges. Acta Biomater 2024; 189:57-72. [PMID: 39424019 DOI: 10.1016/j.actbio.2024.10.021] [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: 07/01/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Spinal cord injury (SCI) is a profound medical condition that significantly hampers motor function, imposing substantial limitations on daily activities and exerting a considerable financial burden on patients and their families. The constrained regenerative capacity of endogenous spinal cord tissue, exacerbated by the inflammatory response following the initial trauma, poses a formidable obstacle to effective therapy. Recent advancements in the field, stem cells, biomaterials, and molecular therapy, show promising outcomes. This review provides a comprehensive analysis of tissue engineering and regenerative medicine approaches for SCI treatment, including cell transplantation, tissue-engineered construct implantation, and other potential therapeutic strategies. Additionally, it sheds light on preclinical animal studies and recent clinical trials incorporating these modalities, providing a glimpse into the evolving landscape of SCI management. STATEMENT OF SIGNIFICANCE: The investigation into spinal cord injury (SCI) treatments focuses on reducing long-term impacts by targeting scar inhibition and enhancing regeneration through stem cells, with or without growth factors. Induced pluripotent stem cells (iPSCs) show promise for autologous use, with clinical trials confirming their safety. Challenges include low cell viability and difficulty in targeted differentiation. Biomaterial scaffolds hold potential for improving cell viability and integration, and extracellular vesicles (EVs) are emerging as a novel therapy. While EV research is in its early stages, stem cell trials demonstrate safety and potential recovery. Advancing tissue engineering approaches with biomaterial scaffolds is crucial for human trials.
Collapse
Affiliation(s)
- Patrick C Ralph
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| | - Sung-Woo Choi
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States; Department of Orthopedic Surgery, Soonchunhyang University Hospital Seoul, Seoul 04401, Republic of Korea
| | - Min Jung Baek
- Department of Obstetrics and Gynecology, CHA University Bundang Medical Center, Seongnam, Gyeonggi-do 13496, Republic of Korea
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States.
| |
Collapse
|
4
|
Kim JW, Kim J, Lee SM, Rim YA, Sung YC, Nam Y, Kim HJ, Kim H, Jung SI, Lim J, Ju JH. Combination of induced pluripotent stem cell-derived motor neuron progenitor cells with irradiated brain-derived neurotrophic factor over-expressing engineered mesenchymal stem cells enhanced restoration of axonal regeneration in a chronic spinal cord injury rat model. Stem Cell Res Ther 2024; 15:173. [PMID: 38886817 PMCID: PMC11184802 DOI: 10.1186/s13287-024-03770-9] [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: 11/28/2023] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a disease that causes permanent impairment of motor, sensory, and autonomic nervous system functions. Stem cell transplantation for neuron regeneration is a promising strategic treatment for SCI. However, selecting stem cell sources and cell transplantation based on experimental evidence is required. Therefore, this study aimed to investigate the efficacy of combination cell transplantation using the brain-derived neurotrophic factor (BDNF) over-expressing engineered mesenchymal stem cell (BDNF-eMSC) and induced pluripotent stem cell-derived motor neuron progenitor cell (iMNP) in a chronic SCI rat model. METHOD A contusive chronic SCI was induced in Sprague-Dawley rats. At 6 weeks post-injury, BDNF-eMSC and iMNP were transplanted into the lesion site via the intralesional route. At 12 weeks post-injury, differentiation and growth factors were evaluated through immunofluorescence staining and western blot analysis. Motor neuron differentiation and neurite outgrowth were evaluated by co-culturing BDNF-eMSC and iMNP in vitro in 2-dimensional and 3-dimensional. RESULTS Combination cell transplantation in the chronic SCI model improved behavioral recovery more than single-cell transplantation. Additionally, combination cell transplantation enhanced mature motor neuron differentiation and axonal regeneration at the injured spinal cord. Both BDNF-eMSC and iMNP played a critical role in neurite outgrowth and motor neuron maturation via BDNF expression. CONCLUSIONS Our results suggest that the combined transplantation of BDNF- eMSC and iMNP in chronic SCI results in a significant clinical recovery. The transplanted iMNP cells predominantly differentiated into mature motor neurons. Additionally, BDNF-eMSC exerts a paracrine effect on neuron regeneration through BDNF expression in the injured spinal cord.
Collapse
Affiliation(s)
- Jang-Woon Kim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Juryun Kim
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Yoojun Nam
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Hyewon Kim
- YiPSCELL, Inc., Seoul, Republic of Korea
| | - Se In Jung
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jooyoung Lim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
- YiPSCELL, Inc., Seoul, Republic of Korea.
| |
Collapse
|
5
|
Wang Z, Li J, Xu T, Guo B, Xie Z, Li M. The Efficacy of Different Material Scaffold-Guided Cell Transplantation in the Treatment of Spinal Cord Injury in Rats: A Systematic Review and Network Meta-analysis. Cell Mol Neurobiol 2024; 44:43. [PMID: 38703332 PMCID: PMC11069479 DOI: 10.1007/s10571-024-01465-6] [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: 12/17/2023] [Accepted: 02/23/2024] [Indexed: 05/06/2024]
Abstract
Cell transplantation is a promising treatment option for spinal cord injury (SCI). However, there is no consensus on the choice of carrier scaffolds to host the cells. This study aims to evaluate the efficacy of different material scaffold-mediated cell transplantation in treating SCI in rats. According to PRISMA's principle, Embase, PubMed, Web of Science, and Cochrane databases were searched, and relevant literature was referenced. Only original research on cell transplantation plus natural or synthetic scaffolds in SCI rats was included. Direct and indirect evidence for improving hind limb motor function was pooled through meta-analysis. A subgroup analysis of some factors that may affect the therapeutic effect was conducted to understand the results fully. In total, 25 studies met the inclusion criteria, in which 293 rats received sham surgery, 78 rats received synthetic material scaffolds, and 219 rats received natural materials scaffolds. The network meta-analysis demonstrated that although synthetic scaffolds were slightly inferior to natural scaffolds in terms of restoring motor function in cell transplantation of SCI rats, no statistical differences were observed between the two (MD: -0.35; 95% CI -2.6 to 1.9). Moreover, the subgroup analysis revealed that the type and number of cells may be important factors in therapeutic efficacy (P < 0.01). Natural scaffolds and synthetic scaffolds are equally effective in cell transplantation of SCI rats without significant differences. In the future, the findings need to be validated in multicenter, large-scale, randomized controlled trials in clinical practice. Trial registration: Registration ID CRD42024459674 (PROSPERO).
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Department of the Second Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Tianqi Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China
- Department of the Second Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Boyu Guo
- Department of the First Clinical Medical College of Nanchang University, No.460, BaYi Street, Nanchang, 330006, Jiangxi Province, China
| | - Zhiping Xie
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No.152 Aiguo Road, Nanchang, 330006, Jiangxi Province, China.
- Department of Neurosurgery, Xiangya Hospital Jiangxi Hospital, Central South University, Nanchang, Jiangxi Province, China.
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No.17, Yongwai Street, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
6
|
Kim JW, Kim J, Mo H, Han H, Rim YA, Ju JH. Stepwise combined cell transplantation using mesenchymal stem cells and induced pluripotent stem cell-derived motor neuron progenitor cells in spinal cord injury. Stem Cell Res Ther 2024; 15:114. [PMID: 38650015 PMCID: PMC11036722 DOI: 10.1186/s13287-024-03714-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is an intractable neurological disease in which functions cannot be permanently restored due to nerve damage. Stem cell therapy is a promising strategy for neuroregeneration after SCI. However, experimental evidence of its therapeutic effect in SCI is lacking. This study aimed to investigate the efficacy of transplanted cells using stepwise combined cell therapy with human mesenchymal stem cells (hMSC) and induced pluripotent stem cell (iPSC)-derived motor neuron progenitor cells (iMNP) in a rat model of SCI. METHODS A contusive SCI model was developed in Sprague-Dawley rats using multicenter animal spinal cord injury study (MASCIS) impactor. Three protocols were designed and conducted as follows: (Subtopic 1) chronic SCI + iMNP, (Subtopic 2) acute SCI + multiple hMSC injections, and (Main topic) chronic SCI + stepwise combined cell therapy using multiple preemptive hMSC and iMNP. Neurite outgrowth was induced by coculturing hMSC and iPSC-derived motor neuron (iMN) on both two-dimensional (2D) and three-dimensional (3D) spheroid platforms during mature iMN differentiation in vitro. RESULTS Stepwise combined cell therapy promoted mature motor neuron differentiation and axonal regeneration at the lesional site. In addition, stepwise combined cell therapy improved behavioral recovery and was more effective than single cell therapy alone. In vitro results showed that hMSC and iMN act synergistically and play a critical role in the induction of neurite outgrowth during iMN differentiation and maturation. CONCLUSIONS Our findings show that stepwise combined cell therapy can induce alterations in the microenvironment for effective cell therapy in SCI. The in vitro results suggest that co-culturing hMSC and iMN can synergistically promote induction of MN neurite outgrowth.
Collapse
Affiliation(s)
- Jang-Woon Kim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | | | - Hyunkyung Mo
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Heeju Han
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- YiPSCELL, Inc, Seoul, South Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Ziai Y, Lanzi M, Rinoldi C, Zargarian SS, Zakrzewska A, Kosik-Kozioł A, Nakielski P, Pierini F. Developing strategies to optimize the anchorage between electrospun nanofibers and hydrogels for multi-layered plasmonic biomaterials. NANOSCALE ADVANCES 2024; 6:1246-1258. [PMID: 38356619 PMCID: PMC10863722 DOI: 10.1039/d3na01022h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/28/2024] [Indexed: 02/16/2024]
Abstract
Polycaprolactone (PCL), a recognized biopolymer, has emerged as a prominent choice for diverse biomedical endeavors due to its good mechanical properties, exceptional biocompatibility, and tunable properties. These attributes render PCL a suitable alternative biomaterial to use in biofabrication, especially the electrospinning technique, facilitating the production of nanofibers with varied dimensions and functionalities. However, the inherent hydrophobicity of PCL nanofibers can pose limitations. Conversely, acrylamide-based hydrogels, characterized by their interconnected porosity, significant water retention, and responsive behavior, present an ideal matrix for numerous biomedical applications. By merging these two materials, one can harness their collective strengths while potentially mitigating individual limitations. A robust interface and effective anchorage during the composite fabrication are pivotal for the optimal performance of the nanoplatforms. Nanoplatforms are subject to varying degrees of tension and physical alterations depending on their specific applications. This is particularly pertinent in the case of layered nanostructures, which require careful consideration to maintain structural stability and functional integrity in their intended applications. In this study, we delve into the influence of the fiber dimensions, orientation and surface modifications of the nanofibrous layer and the hydrogel layer's crosslinking density on their intralayer interface to determine the optimal approach. Comprehensive mechanical pull-out tests offer insights into the interfacial adhesion and anchorage between the layers. Notably, plasma treatment of the hydrophobic nanofibers and the stiffness of the hydrogel layer significantly enhance the mechanical effort required for fiber extraction from the hydrogels, indicating improved anchorage. Furthermore, biocompatibility assessments confirm the potential biomedical applications of the proposed nanoplatforms.
Collapse
Affiliation(s)
- Yasamin Ziai
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| | - Massimiliano Lanzi
- Department of Industrial Chemistry, University of Bologna 40136 Bologna Italy
| | - Chiara Rinoldi
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| | - Anna Zakrzewska
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| | - Alicja Kosik-Kozioł
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| | - Paweł Nakielski
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| | - Filippo Pierini
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research, Polish Academy of Sciences Warsaw 02-106 Poland
| |
Collapse
|
8
|
Wang Y, Ding Y, Guo C. Mesenchymal Stem Cells for the Treatment of Spinal Cord Injury in Rat Models: A Systematic Review and Network Meta-Analysis. Cell Transplant 2024; 33:9636897241262992. [PMID: 38910431 PMCID: PMC11265244 DOI: 10.1177/09636897241262992] [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: 04/09/2024] [Revised: 05/23/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) is one of the hopeful treatments for spinal cord injury (SCI). Most current studies are in animals, and less in humans, and the optimal transplantation strategy for MSCs is still controversial. In this article, we explore the optimal transplantation strategy of MSCs through a network meta-analysis of the effects of MSCs on SCI in animal models. PubMed, Web of Science, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI), Wanfang Database, China Science and Technology Journal Database (VIP), and Chinese Biomedical Literature Service System (SinoMed) databases were searched by computer for randomized controlled studies on MSCs for SCI. Two investigators independently completed the literature screening and data extraction based on the inclusion and exclusion criteria. RevMan 5.4 software was used to assess the quality of the included literature. Stata 16.0 software was used for standard meta-analysis and network meta-analysis. Standardized mean difference (SMD) was used for continuous variables to combine the statistics and calculate 95% confidence interval (95% CI). P < 0.05 was considered a statistically significant difference. Cochrane's Q test and the I2 value were used to indicate the magnitude of heterogeneity. A random-effects model was used if I2 > 50% and P < 0.10 indicated significant heterogeneity between studies, and conversely, a fixed-effects model was used. Evidence network diagrams were drawn based on direct comparisons between various interventions. The surface under the cumulative ranking curve area (SUCRA) was used to predict the ranking of the treatment effects of each intervention. A total of 32 animal studies were included in this article for analysis. The results of the standard meta-analysis showed that MSCs improved motor ability after SCI. The network meta-analysis showed that the best treatment effect was achieved for adipose tissue-derived mesenchymal stromal cells (ADMSCs) in terms of cell source and intrathecal (IT) in terms of transplantation modality. For transplantation timing, the best treatment effect was achieved when transplantation was performed in the subacute phase. The available literature suggests that IT transplantation using ADMSCs in the subacute phase may be the best transplantation strategy to improve functional impairment after SCI. Future high-quality studies are still needed to further validate the results of this study to ensure the reliability of the results.
Collapse
Affiliation(s)
- Yueying Wang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi Ding
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenchen Guo
- Department of Rehabilitation Medicine, Neck, Shoulder, Lumbago and Leg Pain Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
9
|
Zhang D, Sun Y, Liu W. Motor functional recovery efficacy of scaffolds with bone marrow stem cells in rat spinal cord injury: a Bayesian network meta-analysis. Spinal Cord 2023; 61:93-98. [PMID: 35842526 DOI: 10.1038/s41393-022-00836-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN A Bayesian network meta-analysis. OBJECTIVE Spinal cord injury (SCI) can profoundly influence human health and has been linked to lifelong disability. More high-level evidence-based medical research is expected to evaluate the value of stem cells and biomaterial scaffold material therapy for SCI. METHODS We performed a comprehensive search of Web of Science, Cochrane databases, Embase, and PubMed databases. 18 randomized controlled trials including both scaffolds and BMSCs were included. We performed a Bayesian network meta-analysis to compare the motor functional recovery efficacy of different scaffolds with BMSCs in rat SCI. RESULTS In our Bayesian network meta-analysis, the motor functional recovery was found to benefit from scaffolds, BMSCs, and BMSCs combined with scaffolds, but the scaffold and BMSC groups had similar motor functional recovery efficacy, and the BMSCs combined with scaffolds group appeared to show better efficacy than BMSCs and scaffolds alone. Subgroup analysis showed that BMSCs+fibrin, BMSCs+ASC, BMSCs+gelatine, and BMSCs+collagen were the best four treatments for SCI in rat models. CONCLUSIONS These Bayesian network meta-analysis findings strongly indicated that BMSCs combined with scaffolds is more effective to improve motor functional recovery than BMSCs and scaffolds alone. The fibrin, gelatine, ASC, and collagen may be favourable scaffolds for the injured spinal cord and that scaffolds with BMSCs could be a promising option in regeneration therapy for patients with SCI.
Collapse
Affiliation(s)
- Dong Zhang
- Changqing District People's Hospital, Jinan, China
| | - Yifeng Sun
- Department of Orthopedic Surgery, The First Affliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Wei Liu
- Department of Orthopedic Surgery, The First Affliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China.
| |
Collapse
|
10
|
Multiple strategies enhance the efficacy of MSCs transplantation for spinal cord injury. Biomed Pharmacother 2023; 157:114011. [PMID: 36410123 DOI: 10.1016/j.biopha.2022.114011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is a serious complication of the central nervous system (CNS) after spine injury, often resulting in severe sensory, motor, and autonomic dysfunction below the level of injury. To date, there is no effective treatment strategy for SCI. Recently, stem cell therapy has brought hope to patients with neurological diseases. Mesenchymal stem cells (MSCs) are considered to be the most promising source of cellular therapy after SCI due to their immunomodulatory, neuroprotective and angiogenic potential. Considering the limited therapeutic effect of MSCs due to the complex pathophysiological environment following SCI, this paper not only reviews the specific mechanism of MSCs to facilitate SCI repair, but also further discusses the research status of these pluripotent stem cells combined with other therapeutic approaches to promote anatomical and functional recovery post-SCI.
Collapse
|
11
|
Choi YH, Cho SH, Seo J, Ahn JH, Kim YC. Apoptosis Occurs in the Anterior Talofibular Ligament of Patients With Chronic Lateral Ankle Instability: An In Vitro Study. Clin Orthop Relat Res 2022; 480:2420-2429. [PMID: 35973121 PMCID: PMC9653187 DOI: 10.1097/corr.0000000000002337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/06/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Chronic lateral ankle instability is treated operatively, whereas most acute ankle sprains associated with acute anterior talofibular ligament injury are usually treated nonoperatively. This treatment strategy is widely accepted and has been validated using a variety of clinical or radiological methods. We suspected that there may be biological differences between chronic and acutely injured ligaments, particularly with respect to apoptosis. Apoptosis is known to cause ligament degeneration. If it could be demonstrated that apoptosis occurs more in the anterior talofibular ligament tissues of patients with chronic lateral ankle instability compared with patients with acute anterior talofibular ligament injury, biological evidence could be supported. QUESTIONS/PURPOSES We sought to (1) elucidate the difference in the extent of apoptosis between patients with chronic lateral ankle instability and those with acute anterior talofibular ligament injury. In addition, we asked: (2) What is the expression level of apoptotic enzymes such as caspases 3, 7, 8, and 9 and cytochrome c in each patient group? (3) Is there a correlation between apoptotic activities and the symptom duration period of chronic lateral ankle instability? METHODS Between March 2019 and February 2021, 50 patients were prospectively enrolled in this study. Anterior talofibular ligament tissues were harvested from patients who were divided into two groups: the chronic lateral ankle instability group and the acute anterior talofibular ligament injury group. Patients with insufficient remaining ligaments were excluded from the chronic lateral ankle instability group, and cases in which the tissue was severely damaged or the quality of collected tissue was insufficient because of severe impingement into the fracture site were excluded from the acute anterior talofibular ligament injury group. Tissues were collected from 21 patients (11 males and 10 females) in the chronic lateral ankle instability group with a mean age of 37 ± 14 years and from 17 patients (6 males and 11 females) in the acute anterior talofibular ligament injury group with a mean age of 49 ± 17 years. To investigate our first purpose, apoptotic cells were counted using a TUNEL assay. To answer our second question, Western blotting for apoptotic enzymes such as caspases 3, 7, 8, and 9 and cytochrome c was performed to investigate apoptotic activity. Immunohistochemistry was also used to detect apoptotic enzymes. To answer our third question, the time elapsed after the first symptom related to chronic lateral ankle instability occurred and the expression level of each enzyme was investigated. RESULTS More apoptotic cells were observed in the chronic lateral ankle instability group than in the acute anterior talofibular ligament injury group in the TUNEL assay. Western blotting revealed that the apoptotic activities of the chronic lateral ankle instability group were higher than those of the acute anterior talofibular ligament injury group: caspase 3 was 117 in the chronic lateral ankle instability group and 59 in the acute anterior talofibular ligament injury group (mean difference 58 [95% confidence interval (CI) 31 to 86]; p < 0.001), caspase 7 was 138 in the chronic lateral ankle instability group and 45 in the acute anterior talofibular ligament injury group (mean difference 93 [95% CI 58 to 128]; p < 0.001), caspase 8 was 126 in the chronic lateral ankle instability group and 68 in the acute anterior talofibular ligament injury group (mean difference 58 [95% CI 29 to 89]; p < 0.001), caspase 9 was 128 in the chronic lateral ankle instability group and 54 in the acute anterior talofibular ligament injury group (mean difference 74 [95% CI 44 to 104]; p < 0.001), and cytochrome c was 139 in the chronic lateral ankle instability group and 51 in the acute anterior talofibular ligament injury group (mean difference 88 [95% CI 46 to 129]; p < 0.001). Immunohistochemistry revealed higher expression of caspases 3, 7, 8, and 9 and cytochrome c in the chronic lateral ankle instability group compared with those in the acute anterior talofibular ligament injury group. Caspases 3, 7, and 9 showed no correlation with duration of chronic lateral ankle instability symptoms: the Pearson correlation coefficient was 0.22 [95% CI -0.25 to 0.69] for caspase 3 (p = 0.36), 0.29 [95% CI -0.16 to 0.74] for caspase 7 (p = 0.23), and 0.29 [95% CI -0.16 to 0.74] for caspase 9 (p = 0.23). CONCLUSION In chronic lateral ankle instability, apoptotic activity in the anterior talofibular ligament was higher than in acute anterior talofibular ligament injury. CLINICAL RELEVANCE Apoptosis occurs more in chronic injured ligaments than in acutely injured ligaments. Although urgent surgical repair is not required for acute anterior talofibular ligament injury, chronic lateral ankle instability may progress if the nonoperative treatment is not successful. Further research should focus not only on timing of apoptotic progression, but also on biological augmentation to reverse or prevent apoptosis within the anterior talofibular ligament.
Collapse
Affiliation(s)
- Youn-Ho Choi
- Department of Orthopaedic Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hyun Cho
- Department of Orthopaedic Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - JeongYong Seo
- Department of Orthopaedic Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jae Hoon Ahn
- Department of Orthopaedic Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Chung Kim
- Department of Orthopaedic Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
12
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
13
|
Mesenchymal Stem Cell Derived Exosomes Suppress Neuronal Cell Ferroptosis Via lncGm36569/miR-5627-5p/FSP1 Axis in Acute Spinal Cord Injury. Stem Cell Rev Rep 2022; 18:1127-1142. [DOI: 10.1007/s12015-022-10327-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2022] [Indexed: 02/08/2023]
|
14
|
Farid MF, S Abouelela Y, Rizk H. Stem cell treatment trials of spinal cord injuries in animals. Auton Neurosci 2022; 238:102932. [PMID: 35016045 DOI: 10.1016/j.autneu.2021.102932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a serious neurological spinal cord damage that resulted in the loss of temporary or permanent function. However, there are even now no effective therapies for it. So, a new medical promising therapeutic hotspot over the previous decades appeared which was (Stem cell (SC) cure of SCI). Otherwise, animal models are considered in preclinical research as a model for humans to trial a potential new treatment. METHODOLOGY Following articles were saved from different databases (PubMed, Google scholar, Egyptian knowledge bank, Elsevier, Medline, Embase, ProQuest, BMC) on the last two decades, and data were obtained then analyzed. RESULTS This review discusses the type and grading of SCI. As well as different types of stem cells therapy for SCI, including mesenchymal stem cells (MSCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs). The review focuses on the transplantation pathways, clinical evaluation, and clinical signs of different types of SC on different animal models which are summarized in tables to give an easy to reach. CONCLUSION Pharmacological and physiotherapy have limited regenerative power in comparison with stem cells medication in the treatment of SCI. Among several sources of cell therapies, mesenchymal stromal/stem cell (MSC) one is being progressively developed as a trusted important energetic way to repair and regenerate. Finally, a wide-ranged animal models have been condensed that helped in human clinical trial therapies.
Collapse
Affiliation(s)
- Mariam F Farid
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Yara S Abouelela
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Hamdy Rizk
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
15
|
Gu C, Feng J, Waqas A, Deng Y, Zhang Y, Chen W, Long J, Huang S, Chen L. Technological Advances of 3D Scaffold-Based Stem Cell/Exosome Therapy in Tissues and Organs. Front Cell Dev Biol 2021; 9:709204. [PMID: 34568322 PMCID: PMC8458970 DOI: 10.3389/fcell.2021.709204] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, biomaterial scaffolds have been widely applied in the field of tissue engineering and regenerative medicine. Due to different production methods, unique types of three-dimensional (3D) scaffolds can be fabricated to meet the structural characteristics of tissues and organs, and provide suitable 3D microenvironments. The therapeutic effects of stem cell (SC) therapy in tissues and organs are considerable and have attracted the attention of academic researchers worldwide. However, due to the limitations and challenges of SC therapy, exosome therapy can be used for basic research and clinical translation. The review briefly introduces the materials (nature or polymer), shapes (hydrogels, particles and porous solids) and fabrication methods (crosslinking or bioprinting) of 3D scaffolds, and describes the recent progress in SC/exosome therapy with 3D scaffolds over the past 5 years (2016-2020). Normal SC/exosome therapy can improve the structure and function of diseased and damaged tissues and organs. In addition, 3D scaffold-based SC/exosome therapy can significantly improve the structure and function cardiac and neural tissues for the treatment of various refractory diseases. Besides, exosome therapy has the same therapeutic effects as SC therapy but without the disadvantages. Hence, 3D scaffold therapy provides an alternative strategy for treatment of refractory and incurable diseases and has entered a transformation period from basic research into clinical translation as a viable therapeutic option in the future.
Collapse
Affiliation(s)
- Chenyang Gu
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia Feng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- School of Medicine, Southeast University, Nanjing, China
| | - Ahmed Waqas
- School of Medicine, Southeast University, Nanjing, China
| | - Yushu Deng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yifan Zhang
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wanghao Chen
- Department of Neurosurgery, Ninth People Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Long
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shiying Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
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: 38] [Impact Index Per Article: 9.5] [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
|
17
|
Johnson LDV, Pickard MR, Johnson WEB. The Comparative Effects of Mesenchymal Stem Cell Transplantation Therapy for Spinal Cord Injury in Humans and Animal Models: A Systematic Review and Meta-Analysis. BIOLOGY 2021; 10:biology10030230. [PMID: 33809684 PMCID: PMC8001771 DOI: 10.3390/biology10030230] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/28/2021] [Accepted: 03/12/2021] [Indexed: 12/17/2022]
Abstract
Animal models have been used in preclinical research to examine potential new treatments for spinal cord injury (SCI), including mesenchymal stem cell (MSC) transplantation. MSC transplants have been studied in early human trials. Whether the animal models represent the human studies is unclear. This systematic review and meta-analysis has examined the effects of MSC transplants in human and animal studies. Following searches of PubMed, Clinical Trials and the Cochrane Library, published papers were screened, and data were extracted and analysed. MSC transplantation was associated with significantly improved motor and sensory function in humans, and significantly increased locomotor function in animals. However, there are discrepancies between the studies of human participants and animal models, including timing of MSC transplant post-injury and source of MSCs. Additionally, difficulty in the comparison of functional outcome measures across species limits the predictive nature of the animal research. These findings have been summarised, and recommendations for further research are discussed to better enable the translation of animal models to MSC-based human clinical therapy.
Collapse
Affiliation(s)
- Louis D. V. Johnson
- Chester Medical School, University of Chester, Chester CH1 4BJ, UK
- Correspondence: (L.D.V.J.); (W.E.B.J.); Tel.: +44-7557-353206 (L.D.V.J.); +44-774-5616225 (W.E.B.J.)
| | - Mark R. Pickard
- University Centre Shrewsbury, University of Chester, Shrewsbury SY3 8HQ, UK;
| | - William E. B. Johnson
- Chester Medical School, University of Chester, Chester CH1 4BJ, UK
- University Centre Shrewsbury, University of Chester, Shrewsbury SY3 8HQ, UK;
- Correspondence: (L.D.V.J.); (W.E.B.J.); Tel.: +44-7557-353206 (L.D.V.J.); +44-774-5616225 (W.E.B.J.)
| |
Collapse
|
18
|
Ojeda-Hernández DD, Canales-Aguirre AA, Matias-Guiu J, Gomez-Pinedo U, Mateos-Díaz JC. Potential of Chitosan and Its Derivatives for Biomedical Applications in the Central Nervous System. Front Bioeng Biotechnol 2020; 8:389. [PMID: 32432095 PMCID: PMC7214799 DOI: 10.3389/fbioe.2020.00389] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022] Open
Abstract
It is well known that the central nervous system (CNS) has a limited regenerative capacity and that many therapeutic molecules cannot cross the blood brain barrier (BBB). The use of biomaterials has emerged as an alternative to overcome these limitations. For many years, biomedical applications of chitosan have been studied due to its remarkable biological properties, biocompatibility, and high versatility. Moreover, the interest in this biomaterial for CNS biomedical implementation has increased because of its ability to cross the BBB, mucoadhesiveness, and hydrogel formation capacity. Several chitosan-based biomaterials have been applied with promising results as drug, cell and gene delivery vehicles. Moreover, their capacity to form porous scaffolds and to bear cells and biomolecules has offered a way to achieve neural regeneration. Therefore, this review aims to bring together recent works that highlight the potential of chitosan and its derivatives as adequate biomaterials for applications directed toward the CNS. First, an overview of chitosan and its derivatives is provided with an emphasis on the properties that favor different applications. Second, a compilation of works that employ chitosan-based biomaterials for drug delivery, gene therapy, tissue engineering, and regenerative medicine in the CNS is presented. Finally, the most interesting trends and future perspectives of chitosan and its derivatives applications in the CNS are shown.
Collapse
Affiliation(s)
- Doddy Denise Ojeda-Hernández
- Biotecnología Industrial, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Zapopan, Mexico
| | - Alejandro A Canales-Aguirre
- Unidad de Evaluación Preclínica, Biotecnología Médica y Farmacéutica, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | - Jorge Matias-Guiu
- Servicio de Neurología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Ulises Gomez-Pinedo
- Servicio de Neurología, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Juan C Mateos-Díaz
- Biotecnología Industrial, CONACYT Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Zapopan, Mexico
| |
Collapse
|
19
|
Stojanov S, Berlec A. Electrospun Nanofibers as Carriers of Microorganisms, Stem Cells, Proteins, and Nucleic Acids in Therapeutic and Other Applications. Front Bioeng Biotechnol 2020; 8:130. [PMID: 32158751 PMCID: PMC7052008 DOI: 10.3389/fbioe.2020.00130] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/10/2020] [Indexed: 11/13/2022] Open
Abstract
Electrospinning is a technique that uses polymer solutions and strong electric fields to produce nano-sized fibers that have wide-ranging applications. We present here an overview of the use of electrospinning to incorporate biological products into nanofibers, including microorganisms, cells, proteins, and nucleic acids. Although the conditions used during electrospinning limit the already problematic viability/stability of such biological products, their effective incorporation into nanofibers has been shown to be feasible. Synthetic polymers have been more frequently applied to make nanofibers than natural polymers. Polymer blends are commonly used to achieve favorable physical properties of nanofibers. The majority of nanofibers that contain biological product have been designed for therapeutic applications. The incorporation of these biological products into nanofibers can promote their stability or viability, and also allow their delivery to a desired tissue or organ. Other applications include plant protection in agriculture, fermentation in the food industry, biocatalytic environmental remediation, and biosensing. Live cells that have been incorporated into nanofibers include bacteria and fungi. Nanofibers have served as scaffolds for stem cells seeded on a surface, to enable their delivery and application in tissue regeneration and wound healing. Viruses incorporated into nanofibers have been used in gene delivery, as well as in therapies against bacterial infections and cancers. Proteins (hormones, growth factors, and enzymes) and nucleic acids (DNA and RNA) have been incorporated into nanofibers, mainly to treat diseases and enhance their stability. To summarize, incorporation of biological products into nanofibers has numerous advantages, such as providing protection and facilitating controlled delivery from a solid form with a large surface area. Future studies should address the challenge of transferring nanofibers with biological products into practical and industrial use.
Collapse
Affiliation(s)
- Spase Stojanov
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
20
|
Yousefifard M, Nasseri Maleki S, Askarian-Amiri S, Vaccaro AR, Chapman JR, Fehlings MG, Hosseini M, Rahimi-Movaghar V. A combination of mesenchymal stem cells and scaffolds promotes motor functional recovery in spinal cord injury: a systematic review and meta-analysis. J Neurosurg Spine 2020; 32:269-284. [PMID: 31675724 DOI: 10.3171/2019.8.spine19201] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 08/01/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE There is controversy about the role of scaffolds as an adjunctive therapy to mesenchymal stem cell (MSC) transplantation in spinal cord injury (SCI). Thus, the authors aimed to design a meta-analysis on preclinical evidence to evaluate the effectiveness of combination therapy of scaffold + MSC transplantation in comparison with scaffolds alone and MSCs alone in improving motor dysfunction in SCI. METHODS Electronic databases including Medline, Embase, Scopus, and Web of Science were searched from inception until the end of August 2018. Two independent reviewers screened related experimental studies. Animal studies that evaluated the effectiveness of scaffolds and/or MSCs on motor function recovery following experimental SCI were included. The findings were reported as standardized mean difference (SMD) and 95% confidence interval (CI). RESULTS A total of 34 articles were included in the meta-analysis. Analyses show that combination therapy in comparison with the scaffold group alone (SMD 2.00, 95% CI 1.53-2.46, p < 0.0001), the MSCs alone (SMD 1.58, 95% CI 0.84-2.31, p < 0.0001), and the nontreated group (SMD 3.52, 95% CI 2.84-4.20, p < 0.0001) significantly improved motor function recovery. Co-administration of MSCs + scaffolds only in the acute phase of injury (during the first 3 days after injury) leads to a significant recovery compared to scaffold alone (SMD 2.18, p < 0.0001). In addition, the cotransplantation of scaffolds with bone marrow-derived MSCs (SMD 1.99, p < 0.0001) and umbilical cord-derived MSCs (SMD 1.50, p = 0.001) also improved motor function following SCI. CONCLUSIONS The findings showed that scaffolds + MSCs is more effective than scaffolds and MSCs alone in improving motor function following SCI in animal models, when used in the acute phase of injury.
Collapse
Affiliation(s)
- Mahmoud Yousefifard
- 1Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Solmaz Nasseri Maleki
- 1Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alexander R Vaccaro
- 2Department of Orthopedics and Neurosurgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jens R Chapman
- 3Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| | - Michael G Fehlings
- 4Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- 5Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- 6Department of Surgery and Spine Program, University of Toronto, Ontario, Canada
| | - Mostafa Hosseini
- 7Department of Epidemiology and Biostatistics, School of Public Health, and
| | - Vafa Rahimi-Movaghar
- 8Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran; and
- 9Brain and Spinal Injuries Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Hu X, Zhou X, Li Y, Jin Q, Tang W, Chen Q, Aili D, Qian H. Application of stem cells and chitosan in the repair of spinal cord injury. Int J Dev Neurosci 2019; 76:80-85. [PMID: 31302172 DOI: 10.1016/j.ijdevneu.2019.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/25/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022] Open
Abstract
Cytology and histology obstacles have been the main barriers to multiple tissues injury repair. In search of the most promising treatment strategies for spinal cord injury (SCI), stem cell-based transplantation coupled with various materials/technologies have been explored extensively to enhance SCI repair. Chitosan (CS) has demonstrated immense potential for widespread application in the form of scaffolds and micro-particles for SCI repair. The current review summarizes the evidences for stem cell-based transplantation and CS in SCI repair. Stem cells transplantation, which plays a key role in the repair of SCI, mainly results from its neural differentiation potential and neurotrophic effects. Application of CS enhances the survival of grafted stem cells, upregulates the expression level of neurotrophic factors and heightens the neural differentiation of stem cells as well as the functional recovery of spinal cord. Meanwhile, CS can also be exploited as growth factors/RNA carriers to control the release of regenerating molecules which are beneficial to damage spinal cord repair.
Collapse
Affiliation(s)
- Xinyuan Hu
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Xinru Zhou
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Yang Li
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Qian Jin
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Wenjuan Tang
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Qun Chen
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Dilhumar Aili
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Hui Qian
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, Zhenjiang, Jiangsu, People's Republic of China.,Key Laboratory of Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| |
Collapse
|
22
|
Boido M, Ghibaudi M, Gentile P, Favaro E, Fusaro R, Tonda-Turo C. Chitosan-based hydrogel to support the paracrine activity of mesenchymal stem cells in spinal cord injury treatment. Sci Rep 2019; 9:6402. [PMID: 31024032 PMCID: PMC6483991 DOI: 10.1038/s41598-019-42848-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/10/2019] [Indexed: 12/21/2022] Open
Abstract
Advanced therapies which combine cells with biomaterial-based carriers are recognized as an emerging and powerful method to treat challenging diseases, such as spinal cord injury (SCI). By enhancing transplanted cell survival and grafting, biomimetic hydrogels can be properly engineered to encapsulate cells and locate them at the injured site in a minimally invasive way. In this work, chitosan (CS) based hydrogels were developed to host mesenchymal stem cells (MSCs), since their paracrine action can therapeutically enhance the SC regeneration, limiting the formation of a glial scar and reducing cell death at the injured site. An injectable and highly permeable CS-based hydrogel was fabricated having a rapid gelation upon temperature increase from 0 to 37 °C. CS was selected as former material both for its high biocompatibility that guarantees the proper environment for MSCs survival and for its ability to provide anti-inflammatory and anti-oxidant cues. MSCs were mixed with the hydrogel solution prior to gelation. MSC viability was not affected by the CS hydrogel and encapsulated MSCs were able to release MSC-vesicles as well as to maintain their anti-oxidant features. Finally, preliminary in vivo tests on SCI mice revealed good handling of the CS solution loading MSCs during implantation and high encapsulated MSCs survival after 7 days.
Collapse
Affiliation(s)
- M Boido
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Orbassano, 10043, Italy
| | - M Ghibaudi
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Orbassano, 10043, Italy
| | - P Gentile
- School of Engineering, Newcastle University, Newcastle Upon Tyne, NE1 7RU, United Kingdom
| | - E Favaro
- Department of Medical Science, University of Turin, Torino, 10126, Italy
| | - R Fusaro
- Department of Mechanical and Aerospace Engineering - PolitoBIOMed Lab, Politecnico of Torino, Torino, 10129, Italy
| | - C Tonda-Turo
- Department of Mechanical and Aerospace Engineering - PolitoBIOMed Lab, Politecnico of Torino, Torino, 10129, Italy.
| |
Collapse
|
23
|
Sun L, Wang F, Chen H, Liu D, Qu T, Li X, Xu D, Liu F, Yin Z, Chen Y. Co-Transplantation of Human Umbilical Cord Mesenchymal Stem Cells and Human Neural Stem Cells Improves the Outcome in Rats with Spinal Cord Injury. Cell Transplant 2019; 28:893-906. [PMID: 31012325 PMCID: PMC6719499 DOI: 10.1177/0963689719844525] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neural stem cells (NSCs) and mesenchymal stem cells (MSCs) are promising graft materials for cell therapies in spinal cord injury (SCI) models. Previous studies have demonstrated that MSCs can regulate the microenvironment of NSCs and promote their survival rate. Furthermore, several studies indicate that MSCs can reduce stem cell transplantation-linked tumor formation. To our knowledge, no previous studies have determined whether co-transplantation of human umbilical cord mesenchymal stem cells (hUC-MSCs) and human neural stem cells (hNSCs) could improve the outcome in rats with SCI. Therefore, we investigated whether the transplantation of hUC-MSCs combined with hNSCs through an intramedullary injection can improve the outcome of rats with SCI, and explored the underlying mechanisms. In this study, a moderate spinal cord contusion model was established in adult female Wistar rats using an NYU impactor. In total, 108 spinal cord-injured rats were randomly selected and divided into the following five groups: 1) hUC-MSCs group, 2) hNSCs group, 3) hUC-MSCs+hNSCs group, 4) PBS (control) group, and 5) a Sham group. Basso, Beattie and Bresnahan (BBB) behavioral test scores were used to evaluate the motor function of all animals before and after the SCI weekly through the 8th week. Two weeks after transplantation, some rats were sacrificed, immunofluorescence and immunohistochemistry were performed to evaluate the survival and differentiation of the transplanted stem cells, and brain-derived neurotrophic factor (BDNF) was detected by ELISA in the injured spinal cords. At the end of the experiment, we evaluated the remaining myelin sheath and anterior horn neurons in the injured spinal cords using Luxol Fast Blue (LFB) staining. Our results demonstrated that the surviving stem cells in the hUC-MSCs+hNSCs group were significantly increased compared with those in the hUC-MSCs alone and the hNSCs alone groups 2 weeks post-transplantation. Furthermore, the results of the BBB scores and the remaining myelin sheath evaluated via LFB staining in the injured spinal cords demonstrated that the most significantly improved outcome occurred in the hUC-MSCs+hNSCs group. The hUC-MSCs alone and the hNSCs alone groups also had a better outcome compared with that of the PBS-treated group. In conclusion, the present study demonstrates that local intramedullary subacute transplantation of hUC-MSCs, hNSCs, or hUC-MSCs+hNSCs significantly improves the outcome in an in vivo moderate contusion SCI model, and that co-transplantation of hUC-MSCs and hNSCs displayed the best outcome in our experiment.
Collapse
Affiliation(s)
- Lei Sun
- 1 Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, China.,2 Department of Orthopaedics, Taian City Central Hospital, Shandong, China.,3 Department of Orthopaedics, Qian Fo Shan Hospital, Shandong University, Jinan, China
| | - Fan Wang
- 3 Department of Orthopaedics, Qian Fo Shan Hospital, Shandong University, Jinan, China
| | - Heng Chen
- 4 R&D, Cell and Tissue Bank of Shandong Province, Jinan, China
| | - Dong Liu
- 4 R&D, Cell and Tissue Bank of Shandong Province, Jinan, China
| | - Tingyu Qu
- 5 Department of Psychiatry, College of Medicine, University of Illinois at Chicago, USA
| | - Xiaofeng Li
- 1 Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, China
| | - Daxia Xu
- 1 Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, China
| | - Feng Liu
- 2 Department of Orthopaedics, Taian City Central Hospital, Shandong, China
| | - Zhanmin Yin
- 2 Department of Orthopaedics, Taian City Central Hospital, Shandong, China
| | - Yunzhen Chen
- 1 Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
24
|
Ibarra A, Mendieta-Arbesú E, Suarez-Meade P, García-Vences E, Martiñón S, Rodriguez-Barrera R, Lomelí J, Flores-Romero A, Silva-García R, Buzoianu-Anguiano V, Borlongan CV, Frydman TD. Motor Recovery after Chronic Spinal Cord Transection in Rats: A Proof-of-Concept Study Evaluating a Combined Strategy. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:52-62. [DOI: 10.2174/1871527317666181105101756] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/20/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Background:
The chronic phase of Spinal Cord (SC) injury is characterized by the presence
of a hostile microenvironment that causes low activity and a progressive decline in neurological function;
this phase is non-compatible with regeneration. Several treatment strategies have been investigated
in chronic SC injury with no satisfactory results. OBJECTIVE- In this proof-of-concept study,
we designed a combination therapy (Comb Tx) consisting of surgical glial scar removal plus scar inhibition,
accompanied with implantation of mesenchymal stem cells (MSC), and immunization with
neural-derived peptides (INDP).
Methods:
This study was divided into three subsets, all in which Sprague Dawley rats were subjected
to a complete SC transection. Sixty days after injury, animals were randomly allocated into two groups
for therapeutic intervention: control group and animals receiving the Comb-Tx. Sixty-three days after
treatment we carried out experiments analyzing motor recovery, presence of somatosensory evoked
potentials, neural regeneration-related genes, and histological evaluation of serotoninergic fibers.
Results:
Comb-Tx induced a significant locomotor and electrophysiological recovery. An increase in the
expression of regeneration-associated genes and the percentage of 5-HT+ fibers was noted at the caudal
stump of the SC of animals receiving the Comb-Tx. There was a significant correlation of locomotor recovery
with positive electrophysiological activity, expression of GAP43, and percentage of 5-HT+ fibers.
Conclusion:
Comb-Tx promotes motor and electrophysiological recovery in the chronic phase of SC
injury subsequent to a complete transection. Likewise, it is capable of inducing the permissive microenvironment
to promote axonal regeneration.
Collapse
Affiliation(s)
- Antonio Ibarra
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Erika Mendieta-Arbesú
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Paola Suarez-Meade
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Elisa García-Vences
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | | | - Roxana Rodriguez-Barrera
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | - Joel Lomelí
- Instituto Politecnico Nacional, Escuela Superior de Medicina, Ciudad de Mexico, Mexico
| | - Adrian Flores-Romero
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| | | | | | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, United States
| | - Tamara D. Frydman
- Centro de Investigacion en Ciencias de la Salud (CICSA), Universidad Anahuac Mexico Campus Norte, Huixquilucan Estado de Mexico, Mexico
| |
Collapse
|
25
|
Wang J, Zou W, Ma J, Liu J. Biomaterials and Gene Manipulation in Stem Cell-Based Therapies for Spinal Cord Injury. Stem Cells Dev 2019; 28:239-257. [PMID: 30489226 DOI: 10.1089/scd.2018.0169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Spinal cord injury (SCI), a prominent health issue, represents a substantial portion of the global health care burden. Stem cell-based therapies provide novel solutions for SCI treatment, yet obstacles remain in the form of low survival rate, uncontrolled differentiation, and functional recovery. The application of engineered biomaterials in stem cell therapy provides a physicochemical microenvironment that mimics the stem cell niche, facilitating self-renewal, stem cell differentiation, and tissue reorganization. Nonetheless, external microenvironment support is inadequate, and some obstacles persist, for example, limited sources, gradual aging, and immunogenicity of stem cells. Targeted stem cell gene manipulation could eliminate many of these drawbacks, allowing safer, more effective use under regulation of intrinsic mechanisms. Additionally, through genetic labeling of stem cells, their role in tissue engineering may be elucidated. Therefore, combining stem cell therapy, materials science, and genetic modification technologies may shed light on SCI treatment. Herein, recent advances and advantages of biomaterials and gene manipulation, especially with respect to stem cell-based therapies, are highlighted, and their joint performance in SCI is evaluated. Current technological limitations and perspectives on future directions are then discussed. Although this combination is still in the early stages of development, it is highly likely to substantially contribute to stem cell-based therapies in the foreseeable future.
Collapse
Affiliation(s)
- Jiayi Wang
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Zou
- 3 College of Life Sciences, Liaoning Normal University, Dalian, China.,4 Liaoning Key Laboratories of Biotechnology and Molecular Drug Research & Development, Dalian, China
| | - Jingyun Ma
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Liu
- 1 Regenerative Medicine Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China.,2 Stem Cell Clinical Research Center, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
26
|
Improved delivery of PLGA microparticles and microparticle-cell scaffolds in clinical needle gauges using modified viscosity formulations. Int J Pharm 2018; 546:272-278. [PMID: 29753905 DOI: 10.1016/j.ijpharm.2018.05.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 01/06/2023]
Abstract
Polymer microparticles are widely used as acellular drug delivery platforms in regenerative medicine, and have emerging potential as cellular scaffolds for therapeutic cell delivery. In the clinic, PLGA microparticles are typically administered intramuscularly or subcutaneously, with the clinician and clinical application site determining the precise needle gauge used for delivery. Here, we explored the role of needle diameter in microparticle delivery yield, and develop a modified viscosity formulation to improve microparticle delivery across a range of clinically relevant needle diameters. We have identified an optimal biocompatible formulation containing 0.25% pluronic F127 and 0.25% carboxymethyl cellulose, which can increase delivery payload to 520% across needle gauges 21-30G, and note that needle diameter impacts delivery efficacy. We use this formulation to increase the delivery yield of PLGA microparticles, and separately, PLGA-cell scaffolds supporting viable mesenchymal stem cells (MSCs), demonstrating the first in vitro delivery of this cell scaffold system. Together, these results highlight an optimal formulation for the delivery of microparticle and microparticle-cell scaffolds, and illustrate how careful choice of delivery formulation and needle size can dramatically impact delivery payload.
Collapse
|
27
|
Biomaterial Scaffolds in Regenerative Therapy of the Central Nervous System. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7848901. [PMID: 29805977 PMCID: PMC5899851 DOI: 10.1155/2018/7848901] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 02/18/2018] [Accepted: 02/21/2018] [Indexed: 02/08/2023]
Abstract
The central nervous system (CNS) is the most important section of the nervous system as it regulates the function of various organs. Injury to the CNS causes impairment of neurological functions in corresponding sites and further leads to long-term patient disability. CNS regeneration is difficult because of its poor response to treatment and, to date, no effective therapies have been found to rectify CNS injuries. Biomaterial scaffolds have been applied with promising results in regeneration medicine. They also show great potential in CNS regeneration for tissue repair and functional recovery. Biomaterial scaffolds are applied in CNS regeneration predominantly as hydrogels and biodegradable scaffolds. They can act as cellular supportive scaffolds to facilitate cell infiltration and proliferation. They can also be combined with cell therapy to repair CNS injury. This review discusses the categories and progression of the biomaterial scaffolds that are applied in CNS regeneration.
Collapse
|
28
|
Libro R, Bramanti P, Mazzon E. The combined strategy of mesenchymal stem cells and tissue-engineered scaffolds for spinal cord injury regeneration. Exp Ther Med 2017; 14:3355-3368. [PMID: 29042919 PMCID: PMC5639409 DOI: 10.3892/etm.2017.4939] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/03/2017] [Indexed: 01/02/2023] Open
Abstract
Spinal cord injury (SCI) is a traumatic lesion that can result in the loss of motor or sensory neurons. Stem cell (SC)-based therapies have been demonstrated to promote neuronal regeneration following SCI, by releasing a range of trophic factors that support endogenous repair or by differentiating into neurons, or glial cells in order to replace the damaged cells. However, numerous limitations remain for therapies based on SC transplantion alone, including a low rate of survival/engraftment. Nevertheless, scaffolds are 3-dimentional substrates that have revealed to support cell survival, proliferation and differentiation in vivo, by mimicking a more favorable endogenous microenvironment. A multidisciplinary approach, which combines engineered scaffolds with SCs has been proposed as a promising strategy for encouraging spinal cord regeneration. The present review has focused on the regenerative potential of mesenchymal SCs isolated from different sources and combined with various scaffold types, in preclinical and clinical SCI studies.
Collapse
Affiliation(s)
- Rosaliana Libro
- Department of Experimental Neurology, IRCCS Centro Neurolesi ‘Bonino-Pulejo’, I-98124 Messina, Italy
| | - Placido Bramanti
- Department of Experimental Neurology, IRCCS Centro Neurolesi ‘Bonino-Pulejo’, I-98124 Messina, Italy
| | - Emanuela Mazzon
- Department of Experimental Neurology, IRCCS Centro Neurolesi ‘Bonino-Pulejo’, I-98124 Messina, Italy
| |
Collapse
|
29
|
Faccendini A, Vigani B, Rossi S, Sandri G, Bonferoni MC, Caramella CM, Ferrari F. Nanofiber Scaffolds as Drug Delivery Systems to Bridge Spinal Cord Injury. Pharmaceuticals (Basel) 2017; 10:ph10030063. [PMID: 28678209 PMCID: PMC5620607 DOI: 10.3390/ph10030063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/13/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022] Open
Abstract
The complex pathophysiology of spinal cord injury (SCI) may explain the current lack of an effective therapeutic approach for the regeneration of damaged neuronal cells and the recovery of motor functions. A primary mechanical injury in the spinal cord triggers a cascade of secondary events, which are involved in SCI instauration and progression. The aim of the present review is to provide an overview of the therapeutic neuro-protective and neuro-regenerative approaches, which involve the use of nanofibers as local drug delivery systems. Drugs released by nanofibers aim at preventing the cascade of secondary damage (neuro-protection), whereas nanofibrous structures are intended to re-establish neuronal connectivity through axonal sprouting (neuro-regeneration) promotion, in order to achieve a rapid functional recovery of spinal cord.
Collapse
Affiliation(s)
- Angela Faccendini
- Department of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy.
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy.
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy.
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy.
| | | | | | - Franca Ferrari
- Department of Drug Sciences, University of Pavia, Viale Taramelli, 12, 27100 Pavia, Italy.
| |
Collapse
|
30
|
Mullick M, Venkatesh K, Sen D. d-Alanine 2, Leucine 5 Enkephaline (DADLE)-mediated DOR activation augments human hUCB-BFs viability subjected to oxidative stress via attenuation of the UPR. Stem Cell Res 2017; 22:20-28. [DOI: 10.1016/j.scr.2017.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 01/16/2023] Open
|
31
|
Gnanasegaran N, Govindasamy V, Simon C, Gan QF, Vincent-Chong VK, Mani V, Krishnan Selvarajan K, Subramaniam V, Musa S, Abu Kasim NH. Effect of dental pulp stem cells in MPTP-induced old-aged mice model. Eur J Clin Invest 2017; 47:403-414. [PMID: 28369799 DOI: 10.1111/eci.12753] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/24/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disease caused by the loss of dopaminergic (DA-ergic) neurons in the substantia nigra (SN) and represented as a huge threat to the geriatric population. Cell replacement therapies (CRTs) have been proposed as a promising strategy to slow down or replace neuronal loss. Among the widely available cell sources, dental pulp stem cells (DPSCs) portray as an attractive source primarily due to their neural crest origin, ease of tissue procurement and less ethical hurdles. MATERIALS AND METHODS We first demonstrated the in vitro differentiation ability of DPSCs towards DA-ergic-like cells before evaluating their neuro-protection/neuro-restoration capacities in MPTP-induced mice. Transplantation via intrathecal was performed with behavioural assessments being evaluated every fortnight. Subsequent analysis investigating their immuno-modulatory behaviour was conducted using neuronal and microglial cell lines. RESULTS It was apparent that the behavioural parameters began to improve corresponding to tyrosine hydroxylase (TH), dopamine transporter (DAT) and dopamine decarboxylase (AADC) immunostaining in SN and striatum as early as 8-week post-transplantation (P < 0·05). About 60% restoration of DA-ergic neurons was observed at SN in MPTP-treated mice after 12-week post-transplantation. Similarly, their ability to reduce toxic effects of MPTP (DNA damages, reactive oxygen species and nitric oxide release) and regulate cytokine levels was distinctly noted (P < 0·05) upon exposure in in vitro model. CONCLUSIONS Our results suggest that DPSCs may provide a therapeutic benefit in the old-aged PD mice model and may be explored in stem cell-based CRTs especially in geriatric population as an attempt towards 'personalized medicine'.
Collapse
Affiliation(s)
- Nareshwaran Gnanasegaran
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Vijayendran Govindasamy
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Christopher Simon
- Faculty of Applied Sciences, AIMST University, Semeling, Bedong, Kedah, Malaysia
| | - Quan Fu Gan
- Faculty of Applied Sciences, AIMST University, Semeling, Bedong, Kedah, Malaysia
| | - Vui King Vincent-Chong
- Oral Cancer Research and Coordinating Center (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, Saudi Arabia
| | | | - Vellayan Subramaniam
- Laboratory Animal Facility and Management (LAFAM), Faculty of Pharmacy, UiTM Puncak, Alam Selangor, Malaysia
| | - Sabri Musa
- Department of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Abstract
Spinal cord injury (SCI) represents one of the most complicated and heterogeneous pathological processes of central nervous system (CNS) impairments, which is still beyond functional regeneration. Transplantation of mesenchymal stem cells (MSCs) has been shown to promote the repair of the injured spinal cord tissues in animal models, and therefore, there is much interest in the clinical use of these cells. However, many questions which are essential to improve the therapy effects remain unanswered. For instance, the functional roles and related molecular regulatory mechanisms of MSCs in vivo are not yet completely determined. It is important for transplanted cells to migrate into the injured tissue, to survive and undergo neural differentiation, or to play neural protection roles by various mechanisms after SCI. In this review, we will focus on some of the recent knowledge about the biological behavior and function of MSCs in SCI. Meanwhile, we highlight the function of biomaterials to direct the behavior of MSCs based on our series of work on silk fibroin biomaterials and attempt to emphasize combinational strategies such as tissue engineering for functional improvement of SCI.
Collapse
|
33
|
Multichannel polymer scaffold seeded with activated Schwann cells and bone mesenchymal stem cells improves axonal regeneration and functional recovery after rat spinal cord injury. Acta Pharmacol Sin 2017; 38:623-637. [PMID: 28392569 DOI: 10.1038/aps.2017.11] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
The adult mammalian CNS has a limited capacity to regenerate after traumatic injury. In this study, a combinatorial strategy to promote axonal regeneration and functional recovery after spinal cord injury (SCI) was evaluated in adult rats. The rats were subjected to a complete transection in the thoracic spinal cord, and multichannel scaffolds seeded with activated Schwann cells (ASCs) and/or rat bone marrow-derived mesenchymal stem cells (MSCs) were acutely grafted into the 3-mm-wide transection gap. At 4 weeks post-transplantation and thereafter, the rats receiving scaffolds seeded with ASCs and MSCs exhibited significant recovery of nerve function as shown by the Basso, Beattie and Bresnahan (BBB) score and electrophysiological test results. Immunohistochemical analyses at 4 and 8 weeks after transplantation revealed that the implanted MSCs at the lesion/graft site survived and differentiated into neuron-like cells and co-localized with host neurons. Robust bundles of regenerated fibers were identified in the lesion/graft site in the ASC and MSC co-transplantation rats, and neurofilament 200 (NF) staining confirmed that these fibers were axons. Furthermore, myelin basic protein (MBP)-positive myelin sheaths were also identified at the lesion/graft site and confirmed via electron microscopy. In addition to expressing mature neuronal markers, sparse MSC-derived neuron-like cells expressed choline acetyltransferase (ChAT) at the injury site of the ASC and MSC co-transplantation rats. These findings suggest that co-transplantation of ASCs and MSCs in a multichannel polymer scaffold may represent a novel combinatorial strategy for the treatment of spinal cord injury.
Collapse
|