51
|
Regnier TC, Most H. Acupuncture and physical therapy for spinal cord injury: Case report. Explore (NY) 2022:S1550-8307(22)00214-2. [DOI: 10.1016/j.explore.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
|
52
|
Yang R, Pan J, Wang Y, Xia P, Tai M, Jiang Z, Chen G. Application and prospects of somatic cell reprogramming technology for spinal cord injury treatment. Front Cell Neurosci 2022; 16:1005399. [PMID: 36467604 PMCID: PMC9712200 DOI: 10.3389/fncel.2022.1005399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/02/2022] [Indexed: 08/10/2023] Open
Abstract
Spinal cord injury (SCI) is a serious neurological trauma that is challenging to treat. After SCI, many neurons in the injured area die due to necrosis or apoptosis, and astrocytes, oligodendrocytes, microglia and other non-neuronal cells become dysfunctional, hindering the repair of the injured spinal cord. Corrective surgery and biological, physical and pharmacological therapies are commonly used treatment modalities for SCI; however, no current therapeutic strategies can achieve complete recovery. Somatic cell reprogramming is a promising technology that has gradually become a feasible therapeutic approach for repairing the injured spinal cord. This revolutionary technology can reprogram fibroblasts, astrocytes, NG2 cells and neural progenitor cells into neurons or oligodendrocytes for spinal cord repair. In this review, we provide an overview of the transcription factors, genes, microRNAs (miRNAs), small molecules and combinations of these factors that can mediate somatic cell reprogramming to repair the injured spinal cord. Although many challenges and questions related to this technique remain, we believe that the beneficial effect of somatic cell reprogramming provides new ideas for achieving functional recovery after SCI and a direction for the development of treatments for SCI.
Collapse
Affiliation(s)
- Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Yankai Wang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Panhui Xia
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Mingliang Tai
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Zhihao Jiang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
53
|
Qin F, He G, Sun Y, Chen G, Yu Q, Ma X. Overexpression of long non-coding RNA LINC00158 inhibits neuronal apoptosis by promoting autophagy in spinal cord injury. CHINESE J PHYSIOL 2022; 65:282-289. [PMID: 36588354 DOI: 10.4103/0304-4920.360035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Spinal cord injury (SCI) is a common central nervous system disease. It is reported that long non-coding RNA LINC00158 is involved in the process of SCI. The purpose of this study was to explore the biological role of LINC00158 in the SCI. First, we established a rat SCI model by surgical method and evaluated the motor function of rats by the Basso-Beattie-Bresnahan locomotor rating scale. The results showed that the expression of LINC00158 decreased and apoptotic cells increased in the SCI model rats. Meanwhile, we found the upregulated LC3-II/LC3-I, Beclin-1, and p62 in the SCI rats. Then, primary rat spinal cord neurons were exposed to oxygen/glucose deprivation (OGD) as an in vitro cell model of SCI. After OGD treatment, the expression of LINC00158 decreased significantly and the apoptosis of spinal cord neurons increased. OGD treatment resulted in upregulation of LC3-II/LC3-I and Beclin-1 and downregulation of p62 in primary spinal cord neurons, which could be eliminated by overexpression of LINC00158. 3-Methyladenine and chloroquine (autophagy inhibitor) reversed the inhibitory effect of LINC00158 overexpression on apoptosis of primary spinal cord neurons. In conclusion, this study demonstrated that LINC00158 overexpression repressed neuronal apoptosis by promoting autophagy, suggesting that LINC00158 may be a potential therapeutic target in the SCI.
Collapse
Affiliation(s)
- Fuchuang Qin
- Department of Neurosurgery, Shulan (Anji) Hospital, Anji County, Huzhou 313300, Zhejiang, China
| | - Guorong He
- Department of Neurosurgery, Xixi Hospital of Hangzhou, Hangzhou 310023, Zhejiang, China
| | - Yu Sun
- Department of Neurosurgery, Shulan (Anji) Hospital, Anji County, Huzhou 313300, Zhejiang, China
| | - Guangning Chen
- Department of Neurosurgery, Shulan (Anji) Hospital, Anji County, Huzhou 313300, Zhejiang, China
| | - Qijian Yu
- Department of Neurosurgery, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Xilie Ma
- Department of Osteology, Hangzhou Red Cross Hospital, Hangzhou 310004, Zhejiang, China
| |
Collapse
|
54
|
Shang Z, Wang M, Zhang B, Wang X, Wanyan P. Clinical translation of stem cell therapy for spinal cord injury still premature: results from a single-arm meta-analysis based on 62 clinical trials. BMC Med 2022; 20:284. [PMID: 36058903 PMCID: PMC9442938 DOI: 10.1186/s12916-022-02482-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND How much scientific evidence is there to show that stem cell therapy is sufficient in preclinical and clinical studies of spinal cord injury before it is translated into clinical practice? This is a complicated problem. A single, small-sample clinical trial is difficult to answer, and accurate insights into this question can only be given by systematically evaluating all the existing evidence. METHODS The PubMed, Ovid-Embase, Web of Science, and Cochrane databases were searched from inception to February 10, 2022. Two independent reviewers performed the literature search, identified and screened the studies, and performed a quality assessment and data extraction. RESULTS In total, 62 studies involving 2439 patients were included in the analysis. Of these, 42 were single-arm studies, and 20 were controlled studies. The meta-analysis showed that stem cells improved the ASIA impairment scale score by at least one grade in 48.9% [40.8%, 56.9%] of patients with spinal cord injury. Moreover, the rate of improvement in urinary and gastrointestinal system function was 42.1% [27.6%, 57.2%] and 52.0% [23.6%, 79.8%], respectively. However, 28 types of adverse effects were observed to occur due to stem cells and transplantation procedures. Of these, neuropathic pain, abnormal feeling, muscle spasms, vomiting, and urinary tract infection were the most common, with an incidence of > 20%. While no serious adverse effects such as tumorigenesis were reported, this could be due to the insufficient follow-up period. CONCLUSIONS Overall, the results demonstrated that although the efficacy of stem cell therapy is encouraging, the subsequent adverse effects remain concerning. In addition, the clinical trials had problems such as small sample sizes, poor design, and lack of prospective registration, control, and blinding. Therefore, the current evidence is not sufficiently strong to support the clinical translation of stem cell therapy for spinal cord injury, and several problems remain. Additional well-designed animal experiments and high-quality clinical studies are warranted to address these issues.
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Mingchuan Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Baolin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Xin Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China.
- Chengren Institute of Traditional Chinese Medicine, Lanzhou, 730000, Gansu Province, China.
- Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, 730000, China
- The Second Hospital of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
55
|
Lu Y, Zhang W, Tian Z, Liang Q, Liu C, Wu Y, Zhang L, Rong L. The optimal transplantation strategy of umbilical cord mesenchymal stem cells in spinal cord injury: a systematic review and network meta-analysis based on animal studies. Stem Cell Res Ther 2022; 13:441. [PMID: 36056386 PMCID: PMC9438219 DOI: 10.1186/s13287-022-03103-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/31/2022] [Indexed: 12/09/2022] Open
Abstract
Objective Umbilical cord mesenchymal stem cells (UCMSCs) have great potential in the treatment of spinal cord injury. However, the specific therapeutic effect and optimal transplantation strategy are still unclear. Therefore, exploring the optimal treatment strategy of UCMSCs in animal studies by systematic review can provide reference for the development of animal studies and clinical research in the future. Methods Databases of PubMed, Ovid-Embase, Web of Science, CNKI, WanFang, VIP, and CBM were searched for the literature in February 11, 2022. Two independent reviewers performed the literature search, identification, screening, quality assessment, and data extraction. Results and Discussion A total of 40 animal studies were included for combined analysis. In different subgroups, the results of traditional meta-analysis and network meta-analysis were consistent, that is, the therapeutic effect of high-dose (≥ 1 × 106) transplantation of UCMSCs was significantly better than that of low dose (< 1 × 106), the therapeutic effect of local transplantation of UCMSCs was significantly better than that of intravenous transplantation, and the therapeutic effect of subacute transplantation of UCMSCs was significantly better than that of acute and chronic transplantation. However, in view of the inherent risk of bias and limited internal and external validity of the current animal studies, more high-quality, direct comparison studies are needed to further explore the optimal transplantation strategy for UCMSCs in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03103-8.
Collapse
Affiliation(s)
- Yubao Lu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Wei Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Qian Liang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Chenrui Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Yingjie Wu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China.,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China.,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China. .,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No.600 Tianhe Road, Guangzhou, 510630, Guangdong, China. .,National Medical Products Administration (NMPA) Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, 510630, China. .,Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, Guangzhou, 510630, China. .,Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, 510630, China.
| |
Collapse
|
56
|
Tam PKH, Wong KKY, Atala A, Giobbe GG, Booth C, Gruber PJ, Monone M, Rafii S, Rando TA, Vacanti J, Comer CD, Elvassore N, Grikscheit T, de Coppi P. Regenerative medicine: postnatal approaches. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:654-666. [PMID: 35963270 DOI: 10.1016/s2352-4642(22)00193-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Paper 2 of the paediatric regenerative medicine Series focuses on recent advances in postnatal approaches. New gene, cell, and niche-based technologies and their combinations allow structural and functional reconstitution and simulation of complex postnatal cell, tissue, and organ hierarchies. Organoid and tissue engineering advances provide human disease models and novel treatments for both rare paediatric diseases and common diseases affecting all ages, such as COVID-19. Preclinical studies for gastrointestinal disorders are directed towards oesophageal replacement, short bowel syndrome, enteric neuropathy, biliary atresia, and chronic end-stage liver failure. For respiratory diseases, beside the first human tracheal replacement, more complex tissue engineering represents a promising solution to generate transplantable lungs. Genitourinary tissue replacement and expansion usually involve application of biocompatible scaffolds seeded with patient-derived cells. Gene and cell therapy approaches seem appropriate for rare paediatric diseases of the musculoskeletal system such as spinal muscular dystrophy, whereas congenital diseases of complex organs, such as the heart, continue to challenge new frontiers of regenerative medicine.
Collapse
Affiliation(s)
- Paul Kwong Hang Tam
- Faculty of Medicine, Macau University of Science and Technology, Macau Special Administrative Region, China; Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Kenneth Kak Yuen Wong
- Division of Paediatric Surgery, Department of Surgery, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Giovanni Giuseppe Giobbe
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Claire Booth
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Peter J Gruber
- Department of Surgery, Yale University, New Haven, CT, USA
| | - Mimmi Monone
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Shahin Rafii
- Ansary Stem Cell Institute, Department of Medicine, Division of Regenerative Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Thomas A Rando
- Paul F Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph Vacanti
- Department of Pediatric Surgery, Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Mass General Hospital for Children, Boston, MA, USA
| | - Carly D Comer
- Department of Pediatric Surgery, Laboratory for Tissue Engineering and Organ Fabrication, Harvard Medical School, Massachusetts General Hospital, Mass General Hospital for Children, Boston, MA, USA
| | - Nicola Elvassore
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Tracy Grikscheit
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paolo de Coppi
- Stem Cell and Regenerative Medicine Section, Developmental Biology and Cancer Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, UK; Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital, London, UK.
| |
Collapse
|
57
|
Ubiquitin ligase Triad1 promotes neurite outgrowth by inhibiting MDM2-mediated ubiquitination of the neuroprotective factor pleiotropin. J Biol Chem 2022; 298:102443. [PMID: 36055408 PMCID: PMC9531182 DOI: 10.1016/j.jbc.2022.102443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/26/2022] Open
Abstract
Spinal cord injury (SCI) is the most severe result of spine injury, but no effective therapy exists to treat SCI. We have previously shown that the E3 ubiquitin ligase Two RING fingers and DRIL 1 (Triad1) promotes neurite outgrowth after SCI. However, the mechanism by which Triad1 affects neuron growth and the potential involvement of its ubiquitination activity is unclear. Neuroprotective cytokine pleiotrophin (PTN) can promote microglia proliferation and neurotrophic factor secretion to achieve neuroprotection. We find using immunostaining and behavioral assays in rats that the expression of Triad1 and the PTN was peaked at 1 day after SCI and Triad1 improved motor function and histomorphological injury after SCI. We show using flow cytometry and astrocyte/neuronal coculture assays that Triad1 overexpression promoted PTN protein levels, neurotrophic growth factor (NGF) expression, brain-derived neurotrophic factor (BDNF) expression, astrocyte and neuronal viability, and neurite outgrowth but suppressed astrocyte apoptosis, while shRNA-mediated knockdown of Triad1 and PTN had the opposite effects. Ubiquitin ligase murine double mutant 2 (MDM2) has previously been demonstrated to participate in the process of neurite outgrowth and mediate ubiquitination of p53. Furthermore, we demonstrate overexpression of MDM2 downregulated PTN protein levels, NGF expression and BDNF expression in astrocytes, and inhibited neurite outgrowth of neurons. In addition, MDM2 facilitated PTN ubiquitination, which was reversed by Triad1. Finally, we show simultaneous sh-PTN and MDM2 overexpression attenuated the neurite outgrowth-promoting effect of Triad1 overexpression. In conclusion, we propose Triad1 promotes astrocyte-dependent neurite outgrowth to accelerate recovery after SCI by inhibiting MDM2-mediated PTN ubiquitination.
Collapse
|
58
|
Veletić M, Apu EH, Simić M, Bergsland J, Balasingham I, Contag CH, Ashammakhi N. Implants with Sensing Capabilities. Chem Rev 2022; 122:16329-16363. [PMID: 35981266 DOI: 10.1021/acs.chemrev.2c00005] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Because of the aging human population and increased numbers of surgical procedures being performed, there is a growing number of biomedical devices being implanted each year. Although the benefits of implants are significant, there are risks to having foreign materials in the body that may lead to complications that may remain undetectable until a time at which the damage done becomes irreversible. To address this challenge, advances in implantable sensors may enable early detection of even minor changes in the implants or the surrounding tissues and provide early cues for intervention. Therefore, integrating sensors with implants will enable real-time monitoring and lead to improvements in implant function. Sensor integration has been mostly applied to cardiovascular, neural, and orthopedic implants, and advances in combined implant-sensor devices have been significant, yet there are needs still to be addressed. Sensor-integrating implants are still in their infancy; however, some have already made it to the clinic. With an interdisciplinary approach, these sensor-integrating devices will become more efficient, providing clear paths to clinical translation in the future.
Collapse
Affiliation(s)
- Mladen Veletić
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ehsanul Hoque Apu
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Mitar Simić
- Faculty of Electrical Engineering, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Jacob Bergsland
- The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ilangko Balasingham
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
59
|
Agarwal G, Roy A, Kumar H, Srivastava A. Graphene-collagen cryogel controls neuroinflammation and fosters accelerated axonal regeneration in spinal cord injury. BIOMATERIALS ADVANCES 2022; 139:212971. [PMID: 35882128 DOI: 10.1016/j.bioadv.2022.212971] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/11/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
Spinal cord injury (SCI) is a devastating condition resulting in loss of motor function. The pathology of SCI is multifaceted and involves a cascade of events, including neuroinflammation and neuronal degeneration at the epicenter, limiting repair process. We developed a supermacroporous, mechanically elastic, electro-conductive, graphene crosslinked collagen (Gr-Col) cryogels for the regeneration of the spinal cord post-injury. The effects of graphene in controlling astrocytes reactivity and microglia polarization are evaluated in spinal cord slice organotypic culture and rat spinal cord lateral hemisection model of SCI. In our work, the application of external electric stimulus results in the enhanced expression of neuronal markers in an organotypic culture. The implantation of Gr-Col cryogels in rat thoracic T9-T11 hemisection model demonstrates an improved functional recovery within 14 days post-injury (DPI), promoted myelination, and decreases the lesion volume at the injury site. Decrease in the expression of STAT3 in the implanted Gr-Col cryogels may be responsible for the decrease in astrocytes reactivity. Microglia cells within the implanted cryogels shows higher anti-inflammatory phenotype (M2) than inflammatory (M1) phenotype. The higher expression of mature axonal markers like β-tubulin III, GAP43, doublecortin, and neurofilament 200 in the implanted Gr-Col cryogel confirms the axonal regeneration after 28 DPI. Gr-Col cryogels also modulate the production of ECM matrix, favouring the axonal regeneration. This study shows that Gr-Col cryogels decreases neuroinflammation and accelerate axonal regeneration.
Collapse
Affiliation(s)
- Gopal Agarwal
- Department of Biotechnology, National Institute of Pharmaceutical Educational and Research, Ahmedabad, Gandhinagar, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Educational and Research, Ahmedabad, Gandhinagar, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Educational and Research, Ahmedabad, Gandhinagar, India.
| | - Akshay Srivastava
- Department of Medical Device, National Institute of Pharmaceutical Educational and Research, Ahmedabad, Gandhinagar, India.
| |
Collapse
|
60
|
Zarepour A, Bal Öztürk A, Koyuncu Irmak D, Yaşayan G, Gökmen A, Karaöz E, Zarepour A, Zarrabi A, Mostafavi E. Combination Therapy Using Nanomaterials and Stem Cells to Treat Spinal Cord Injuries. Eur J Pharm Biopharm 2022; 177:224-240. [PMID: 35850168 DOI: 10.1016/j.ejpb.2022.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
As a part of the central nervous system, the spinal cord (SC) provides most of the communications between the brain and other parts of the body. Any damage to SC interrupts this communication, leading to serious problems, which may remain for the rest of their life. Due to its significant impact on patients' quality of life and its exorbitant medical costs, SC injury (SCI) is known as one of the most challengeable diseases in the world. Thus, it is critical to introduce highly translatable therapeutic platforms for SCI treatment. So far, different strategies have been introduced, among which utilizing various types of stem cells is one of the most interesting ones. The capability of stem cells to differentiate into several types of cell lines makes them promising candidates for the regeneration of injured tissues. One of the other interesting and novel strategies for SCI treatment is the application of nanomaterials, which could appear as a carrier for therapeutic agents or as a platform for culturing the cells. Combining these two approaches, stem cells and nanomaterials, could provide promising therapeutic strategies for SCI management. Accordingly, in this review we have summarized some of the recent advancements in which the applications of different types of stem cells and nanomaterials, alone and in combination forms, were evaluated for SCI treatment.
Collapse
Affiliation(s)
- Arezou Zarepour
- Radiology Department, Kashan University of Medical Sciences, Kashan, Isfahan, Iran
| | - Ayça Bal Öztürk
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey; Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Zeytinburnu, Turkey
| | | | - Gökçen Yaşayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Aylin Gökmen
- Molecular Biology and Genetics Department, Faculty of Engineering and Natural Sciences, Bahcesehir University, Besiktas, Istanbul, Turkey
| | - Erdal Karaöz
- Liv Hospital, Center for Regenerative Medicine and Stem Cell Manufacturing (LivMedCell), İstanbul, Turkey
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
61
|
Pinelli F, Pizzetti F, Veneruso V, Petillo E, Raghunath M, Perale G, Veglianese P, Rossi F. Biomaterial-Mediated Factor Delivery for Spinal Cord Injury Treatment. Biomedicines 2022; 10:biomedicines10071673. [PMID: 35884981 PMCID: PMC9313204 DOI: 10.3390/biomedicines10071673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 07/05/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is an injurious process that begins with immediate physical damage to the spinal cord and associated tissues during an acute traumatic event. However, the tissue damage expands in both intensity and volume in the subsequent subacute phase. At this stage, numerous events exacerbate the pathological condition, and therein lies the main cause of post-traumatic neural degeneration, which then ends with the chronic phase. In recent years, therapeutic interventions addressing different neurodegenerative mechanisms have been proposed, but have met with limited success when translated into clinical settings. The underlying reasons for this are that the pathogenesis of SCI is a continued multifactorial disease, and the treatment of only one factor is not sufficient to curb neural degeneration and resulting paralysis. Recent advances have led to the development of biomaterials aiming to promote in situ combinatorial strategies using drugs/biomolecules to achieve a maximized multitarget approach. This review provides an overview of single and combinatorial regenerative-factor-based treatments as well as potential delivery options to treat SCIs.
Collapse
Affiliation(s)
- Filippo Pinelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
| | - Fabio Pizzetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
| | - Valeria Veneruso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Emilia Petillo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
| | - Michael Raghunath
- Center for Cell Biology and Tissue Engineering, Institute for Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences (ZHAW), 8820 Wädenswil, Switzerland;
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI), Via Buffi 13, 6900 Lugano, Switzerland;
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Pietro Veglianese
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy;
- Correspondence: (P.V.); (F.R.); Tel.: +39-02-3901-4205 (P.V.); +39-02-2399-3145 (F.R.)
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milan, Italy; (F.P.); (F.P.); (E.P.)
- Correspondence: (P.V.); (F.R.); Tel.: +39-02-3901-4205 (P.V.); +39-02-2399-3145 (F.R.)
| |
Collapse
|
62
|
Nout-Lomas YS. Traumatic Nervous System Injury. Vet Clin North Am Equine Pract 2022; 38:363-377. [PMID: 35810150 DOI: 10.1016/j.cveq.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Mechanisms of traumatic nervous system injury to a degree are similar, but differences exist in etiology, pathophysiology, and treatment of brain, spinal cord, and peripheral nerve injury. The most common clinical abnormalities seen in the horse are abnormal level of consciousness, abnormal behavior, seizures, cranial nerve deficits, vestibular disease, tetra- and paraparesis or paraplegia, cauda equina syndrome, specific gait deficits, and muscle atrophy. Treatments are directed toward reducing inflammation and swelling, halting secondary injury, and promoting mechanisms of neuroregeneration and plasticity. Prognosis depends on the severity of primary injury and the neuroanatomic location and extent of nervous tissue damage.
Collapse
Affiliation(s)
- Yvette S Nout-Lomas
- Department of Clinical Sciences, Johnson Family Equine Hospital, Colorado State University, 2230 Gillette Drive, Fort Collins, CO 80523-1678, USA.
| |
Collapse
|
63
|
Ali M, Namjoshi S, Benson HAE, Mohammed Y, Kumeria T. Dissolvable polymer microneedles for drug delivery and diagnostics. J Control Release 2022; 347:561-589. [PMID: 35525331 DOI: 10.1016/j.jconrel.2022.04.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
Dissolvable transdermal microneedles (μND) are promising micro-devices used to transport a wide selection of active compounds into the skin. To provide an effective therapeutic outcome, μNDs must pierce the human stratum corneum (~10 to 20 μm), without rupturing or bending during penetration, then release their cargo at the predetermined area and time. The ability of dissolvable μND arrays/patches to sufficiently pierce the skin is a crucial requirement, which depends on the material composition, μND geometry and fabrication techniques. This comprehensive review not only provides contemporary knowledge on the μND design approaches, but also the materials science facilitating these delivery systems and the opportunities these advanced materials can provide to enhance clinical outcomes.
Collapse
Affiliation(s)
- Masood Ali
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia
| | - Sarika Namjoshi
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia; Vaxxas Pty Ltd, Brisbane, Woolloongabba, QLD 4102, Australia
| | - Heather A E Benson
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; Basil Hetzel institute for Translational Health Research, Adelaide, SA 5001, Australia.
| | - Yousuf Mohammed
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney. NSW 2052, Australia; Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| |
Collapse
|
64
|
Lu D, Yang Y, Zhang P, Ma Z, Li W, Song Y, Feng H, Yu W, Ren F, Li T, Zeng H, Wang J. Development and Application of Three-Dimensional Bioprinting Scaffold in the Repair of Spinal Cord Injury. Tissue Eng Regen Med 2022; 19:1113-1127. [PMID: 35767151 DOI: 10.1007/s13770-022-00465-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 01/04/2023] Open
Abstract
Spinal cord injury (SCI) is a disabling and destructive central nervous system injury that has not yet been successfully treated at this stage. Three-dimensional (3D) bioprinting has become a promising method to produce more biologically complex microstructures, which fabricate living neural constructs with anatomically accurate complex geometries and spatial distributions of neural stem cells, and this is critical in the treatment of SCI. With the development of 3D printing technology and the deepening of research, neural tissue engineering research using different printing methods, bio-inks, and cells to repair SCI has achieved certain results. Although satisfactory results have not yet been achieved, they have provided novel ideas for the clinical treatment of SCI. Considering the potential impact of 3D bioprinting technology on neural studies, this review focuses on 3D bioprinting methods widely used in SCI neural tissue engineering, and the latest technological applications of bioprinting of nerve tissues for the repair of SCI are discussed. In addition to introducing the recent progress, this work also describes the existing limitations and highlights emerging possibilities and future prospects in this field.
Collapse
Affiliation(s)
- Dezhi Lu
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yang Yang
- Department of Rehabilitation Medicine, Shandong Provincial Third Hospital, Shandong, 250000, China
| | - Pingping Zhang
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yan Song
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Haiyang Feng
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Wenqiang Yu
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Fuchao Ren
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, 261053, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Hong Zeng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
65
|
Ashammakhi N, GhavamiNejad A, Tutar R, Fricker A, Roy I, Chatzistavrou X, Hoque Apu E, Nguyen KL, Ahsan T, Pountos I, Caterson EJ. Highlights on Advancing Frontiers in Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:633-664. [PMID: 34210148 PMCID: PMC9242713 DOI: 10.1089/ten.teb.2021.0012] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/15/2021] [Indexed: 01/05/2023]
Abstract
The field of tissue engineering continues to advance, sometimes in exponential leaps forward, but also sometimes at a rate that does not fulfill the promise that the field imagined a few decades ago. This review is in part a catalog of success in an effort to inform the process of innovation. Tissue engineering has recruited new technologies and developed new methods for engineering tissue constructs that can be used to mitigate or model disease states for study. Key to this antecedent statement is that the scientific effort must be anchored in the needs of a disease state and be working toward a functional product in regenerative medicine. It is this focus on the wildly important ideas coupled with partnered research efforts within both academia and industry that have shown most translational potential. The field continues to thrive and among the most important recent developments are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies that warrant special attention. Developments in the aforementioned areas as well as future directions are highlighted in this article. Although several early efforts have not come to fruition, there are good examples of commercial profitability that merit continued investment in tissue engineering. Impact statement Tissue engineering led to the development of new methods for regenerative medicine and disease models. Among the most important recent developments in tissue engineering are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies. These technologies and an understanding of them will have impact on the success of tissue engineering and its translation to regenerative medicine. Continued investment in tissue engineering will yield products and therapeutics, with both commercial importance and simultaneous disease mitigation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, Michigan, USA
| | - Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Annabelle Fricker
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Ehsanul Hoque Apu
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Taby Ahsan
- RoosterBio, Inc., Frederick, Maryland, USA
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
| | - Edward J. Caterson
- Division of Plastic Surgery, Department of Surgery, Nemours/Alfred I. du Pont Hospital for Children, Wilmington, Delaware, USA
| |
Collapse
|
66
|
Wang H, Xia Y, Li B, Li Y, Fu C. Reverse Adverse Immune Microenvironments by Biomaterials Enhance the Repair of Spinal Cord Injury. Front Bioeng Biotechnol 2022; 10:812340. [PMID: 35646849 PMCID: PMC9136098 DOI: 10.3389/fbioe.2022.812340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/29/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) is a severe and traumatic disorder that ultimately results in the loss of motor, sensory, and autonomic nervous function. After SCI, local immune inflammatory response persists and does not weaken or disappear. The interference of local adverse immune factors after SCI brings great challenges to the repair of SCI. Among them, microglia, macrophages, neutrophils, lymphocytes, astrocytes, and the release of various cytokines, as well as the destruction of the extracellular matrix are mainly involved in the imbalance of the immune microenvironment. Studies have shown that immune remodeling after SCI significantly affects the survival and differentiation of stem cells after transplantation and the prognosis of SCI. Recently, immunological reconstruction strategies based on biomaterials have been widely explored and achieved good results. In this review, we discuss the important factors leading to immune dysfunction after SCI, such as immune cells, cytokines, and the destruction of the extracellular matrix. Additionally, the immunomodulatory strategies based on biomaterials are summarized, and the clinical application prospects of these immune reconstructs are evaluated.
Collapse
|
67
|
Novel Strategies for Spinal Cord Regeneration. Int J Mol Sci 2022; 23:ijms23094552. [PMID: 35562941 PMCID: PMC9102050 DOI: 10.3390/ijms23094552] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 02/04/2023] Open
Abstract
A spinal cord injury (SCI) is one of the most devastating lesions, as it can damage the continuity and conductivity of the central nervous system, resulting in complex pathophysiology. Encouraged by the advances in nanotechnology, stem cell biology, and materials science, researchers have proposed various interdisciplinary approaches for spinal cord regeneration. In this respect, the present review aims to explore the most recent developments in SCI treatment and spinal cord repair. Specifically, it briefly describes the characteristics of SCIs, followed by an extensive discussion on newly developed nanocarriers (e.g., metal-based, polymer-based, liposomes) for spinal cord delivery, relevant biomolecules (e.g., growth factors, exosomes) for SCI treatment, innovative cell therapies, and novel natural and synthetic biomaterial scaffolds for spinal cord regeneration.
Collapse
|
68
|
The Role of Tissue Geometry in Spinal Cord Regeneration. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58040542. [PMID: 35454380 PMCID: PMC9028021 DOI: 10.3390/medicina58040542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022]
Abstract
Unlike peripheral nerves, axonal regeneration is limited following injury to the spinal cord. While there may be reduced regenerative potential of injured neurons, the central nervous system (CNS) white matter environment appears to be more significant in limiting regrowth. Several factors may inhibit regeneration, and their neutralization can modestly enhance regrowth. However, most investigations have not considered the cytoarchitecture of spinal cord white matter. Several lines of investigation demonstrate that axonal regeneration is enhanced by maintaining, repairing, or reconstituting the parallel geometry of the spinal cord white matter. In this review, we focus on environmental factors that have been implicated as putative inhibitors of axonal regeneration and the evidence that their organization may be an important determinant in whether they inhibit or promote regeneration. Consideration of tissue geometry may be important for developing successful strategies to promote spinal cord regeneration.
Collapse
|
69
|
Shang Z, Li D, Chen J, Wang R, Wang M, Zhang B, Wang X, Wanyan P. What Is the Optimal Timing of Transplantation of Neural Stem Cells in Spinal Cord Injury? A Systematic Review and Network Meta-Analysis Based on Animal Studies. Front Immunol 2022; 13:855309. [PMID: 35371014 PMCID: PMC8965614 DOI: 10.3389/fimmu.2022.855309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/15/2022] [Indexed: 01/13/2023] Open
Abstract
Objective The optimal transplantation timing of neural stem cells in spinal cord injury is fully explored in animal studies to reduce the risk of transformation to clinical practice and to provide valuable reference for future animal studies and clinical research. Method Seven electronic databases, namely, PubMed, Web of Science, Embase, Wanfang, Chinese Scientific Journal Database (CSJD-VIP), China Biomedical Literature Database (CBM), and China National Knowledge Infrastructure (CNKI), were searched. The studies were retrieved from inception to November 2021. Two researchers independently screened the literature, extracted data, and evaluated the methodological quality based on the inclusion criteria. Results and Discussion Thirty-nine studies were incorporated into the final analyses. Based on the subgroup of animal models and transplantation dose, the results of network meta-analysis showed that the effect of transplantation in the subacute phase might be the best. However, the results of traditional meta-analysis were inconsistent. In the moderate-dose group of moderate spinal cord injury model and the low-dose group of severe spinal cord injury model, transplantation in the subacute phase did not significantly improve motor function. Given the lack of evidence for direct comparison between different transplantation phases, the indirectness of our network meta-analysis, and the low quality of evidence in current animal studies, our confidence in recommending cell transplantation in the subacute phase is limited. In the future, more high-quality, direct comparative studies are needed to explore this issue in depth.
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Dongliang Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jinlei Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - RuiRui Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Mingchuan Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Xin Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China.,Chengren Institute of Traditional Chinese Medicine, Lanzhou, China.,Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Pingping Wanyan
- Basic Medical College, Gansu University of Chinese Medicine, Lanzhou, China.,Department of Nephrology, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
70
|
Shang Z, Wang R, Li D, Chen J, Zhang B, Wang M, Wang X, Wanyan P. Spinal Cord Injury: A Systematic Review and Network Meta-Analysis of Therapeutic Strategies Based on 15 Types of Stem Cells in Animal Models. Front Pharmacol 2022; 13:819861. [PMID: 35359872 PMCID: PMC8964098 DOI: 10.3389/fphar.2022.819861] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
Objective: The optimal therapeutic strategies of stem cells for spinal cord injury (SCI) are fully explored in animal studies to promote the translation of preclinical findings to clinical practice, also to provide guidance for future animal experiments and clinical studies. Methods: PubMed, Web of Science, Embase, CNKI, Wangfang, VIP, and CBM were searched from inception to September 2021. Screening of search results, data extraction, and references quality evaluation were undertaken independently by two reviewers. Results and Discussion: A total of 188 studies were included for data analysis. Results of traditional meta-analysis showed that all 15 diverse types of stem cells could significantly improve locomotor function of animals with SCI, and results of further network meta-analysis showed that adipose-derived mesenchymal stem cells had the greatest therapeutic potential for SCI. Moreover, a higher dose (≥1 × 106) of stem cell transplantation had better therapeutic effect, transplantation in the subacute phase (3–14 days, excluding 3 days) was the optimal timing, and intralesional transplantation was the optimal route. However, the evidence of current animal studies is of limited quality, and more high-quality research is needed to further explore the optimal therapeutic strategies of stem cells, while the design and implementation of experiments, as well as measurement and reporting of results for animal studies, need to be further improved and standardized to reduce the risk when the results of animal studies are translated to the clinic. Systematic Review Registration: [website], identifier [registration number].
Collapse
Affiliation(s)
- Zhizhong Shang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Ruirui Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Dongliang Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Jinlei Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Mingchuan Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
| | - Xin Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, China
- Chengren Institute of Traditional Chinese Medicine, Lanzhou, China
- Department of Spine, Changzheng Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| | - Pingping Wanyan
- Gansu University of Chinese Medicine, Lanzhou, China
- The Second Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Xin Wang, ; Pingping Wanyan,
| |
Collapse
|
71
|
Van den Bos J, Ouaamari YE, Wouters K, Cools N, Wens I. Are Cell-Based Therapies Safe and Effective in the Treatment of Neurodegenerative Diseases? A Systematic Review with Meta-Analysis. Biomolecules 2022; 12:340. [PMID: 35204840 PMCID: PMC8869169 DOI: 10.3390/biom12020340] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
Over the past two decades, significant advances have been made in the field of regenerative medicine. However, despite being of the utmost clinical urgency, there remains a paucity of therapeutic strategies for conditions with substantial neurodegeneration such as (progressive) multiple sclerosis (MS), spinal cord injury (SCI), Parkinson's disease (PD) and Alzheimer's disease (AD). Different cell types, such as mesenchymal stromal cells (MSC), neuronal stem cells (NSC), olfactory ensheathing cells (OEC), neurons and a variety of others, already demonstrated safety and regenerative or neuroprotective properties in the central nervous system during the preclinical phase. As a result of these promising findings, in recent years, these necessary types of cell therapies have been intensively tested in clinical trials to establish whether these results could be confirmed in patients. However, extensive research is still needed regarding elucidating the exact mechanism of action, possible immune rejection, functionality and survival of the administered cells, dose, frequency and administration route. To summarize the current state of knowledge, we conducted a systematic review with meta-analysis. A total of 27,043 records were reviewed by two independent assessors and 71 records were included in the final quantitative analysis. These results show that the overall frequency of serious adverse events was low: 0.03 (95% CI: 0.01-0.08). In addition, several trials in MS and SCI reported efficacy data, demonstrating some promising results on clinical outcomes. All randomized controlled studies were at a low risk of bias due to appropriate blinding of the treatment, including assessors and patients. In conclusion, cell-based therapies in neurodegenerative disease are safe and feasible while showing promising clinical improvements. Nevertheless, given their high heterogeneity, the results require a cautious approach. We advocate for the harmonization of study protocols of trials investigating cell-based therapies in neurodegenerative diseases, adverse event reporting and investigation of clinical outcomes.
Collapse
Affiliation(s)
- Jasper Van den Bos
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| | - Yousra El Ouaamari
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| | - Kristien Wouters
- Clinical Trial Center (CTC), CRC Antwerp, Antwerp University Hospital, University of Antwerp, Drie Eikenstraat 655, B-2650 Edegem, Belgium;
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
- Center for Cell Therapy and Regenerative Medicine (CCRG), Antwerp University Hospital, Drie Eikenstraat 655, B-2650 Edegem, Belgium
| | - Inez Wens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| |
Collapse
|
72
|
Suzuki H, Imajo Y, Funaba M, Nishida N, Sakamoto T, Sakai T. Current Concepts of Neural Stem/Progenitor Cell Therapy for Chronic Spinal Cord Injury. Front Cell Neurosci 2022; 15:794692. [PMID: 35185471 PMCID: PMC8850278 DOI: 10.3389/fncel.2021.794692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic spinal cord injury (SCI) is a devastating condition that results in major neurological deficits and social burden. It continues to be managed symptomatically, and no real therapeutic strategies have been devised for its treatment. Neural stem/neural progenitor cells (NSCs/NPCs) being used for the treatment of chronic SCI in experimental SCI models can not only replace the lost cells and remyelinate axons in the injury site but also support their growth and provide neuroprotective factors. Currently, several clinical studies using NSCs/NPCs are underway worldwide. NSCs/NPCs also have the potential to differentiate into all three neuroglial lineages to regenerate neural circuits, demyelinate denuded axons, and provide trophic support to endogenous cells. This article explains the challenging pathophysiology of chronic SCI and discusses key NSC/NPC-based techniques having the greatest potential for translation over the next decade.
Collapse
|
73
|
Kiyotake EA, Martin MD, Detamore MS. Regenerative rehabilitation with conductive biomaterials for spinal cord injury. Acta Biomater 2022; 139:43-64. [PMID: 33326879 DOI: 10.1016/j.actbio.2020.12.021] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
The individual approaches of regenerative medicine efforts alone and rehabilitation efforts alone have not yet fully restored function after severe spinal cord injury (SCI). Regenerative rehabilitation may be leveraged to promote regeneration of the spinal cord tissue, and promote reorganization of the regenerated neural pathways and intact spinal circuits for better functional recovery for SCI. Conductive biomaterials may be a linchpin that empowers the synergy between regenerative medicine and rehabilitation approaches, as electrical stimulation applied to the spinal cord could facilitate neural reorganization. In this review, we discuss current regenerative medicine approaches in clinical trials and the rehabilitation, or neuromodulation, approaches for SCI, along with their respective translational limitations. Furthermore, we review the translational potential, in a surgical context, of conductive biomaterials (e.g., conductive polymers, carbon-based materials, metallic nanoparticle-based materials) as they pertain to SCI. While pre-formed scaffolds may be difficult to translate to human contusion SCIs, injectable composites that contain blended conductive components and can form within the injury may be more translational. However, given that there are currently no in vivo SCI studies that evaluated conductive materials combined with rehabilitation approaches, we discuss several limitations of conductive biomaterials, including demonstrating safety and efficacy, that will need to be addressed in the future for conductive biomaterials to become SCI therapeutics. Even so, the use of conductive biomaterials creates a synergistic opportunity to merge the fields of regenerative medicine and rehabilitation and redefine what regenerative rehabilitation means for the spinal cord. STATEMENT OF SIGNIFICANCE: For spinal cord injury (SCI), the individual approaches of regenerative medicine and rehabilitation are insufficient to fully restore functional recovery; however, the goal of regenerative rehabilitation is to combine these two disparate fields to maximize the functional outcomes. Concepts similar to regenerative rehabilitation for SCI have been discussed in several reviews, but for the first time, this review considers how conductive biomaterials may synergize the two approaches. We cover current regenerative medicine and rehabilitation approaches for SCI, and the translational advantages and disadvantages, in a surgical context, of conductive biomaterials used in biomedical applications that may be additionally applied to SCI. Furthermore, we identify the current limitations and translational challenges for conductive biomaterials before they may become therapeutics for SCI.
Collapse
|
74
|
Gharooni AA, Kwon BK, Fehlings MG, Boerger TF, Rodrigues-Pinto R, Koljonen PA, Kurpad SN, Harrop JS, Aarabi B, Rahimi-Movaghar V, Wilson JR, Davies BM, Kotter MRN, Guest JD. Developing Novel Therapies for Degenerative Cervical Myelopathy [AO Spine RECODE-DCM Research Priority Number 7]: Opportunities From Restorative Neurobiology. Global Spine J 2022; 12:109S-121S. [PMID: 35174725 PMCID: PMC8859698 DOI: 10.1177/21925682211052920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY DESIGN Narrative review. OBJECTIVES To provide an overview of contemporary therapies for the James Lind Alliance priority setting partnership for degenerative cervical myelopathy (DCM) question: 'Can novel therapies, including stem-cell, gene, pharmacological and neuroprotective therapies, be identified to improve the health and wellbeing of people living with DCM and slow down disease progression?' METHODS A review of the literature was conducted to outline the pathophysiology of DCM and present contemporary therapies that may hold therapeutic value in 3 broad categories of neuroprotection, neuroregeneration, and neuromodulation. RESULTS Chronic spinal cord compression leads to ischaemia, neuroinflammation, demyelination, and neuronal loss. Surgical intervention may halt progression and improve symptoms, though the majority do not make a full recovery leading to lifelong disability. Neuroprotective agents disrupt deleterious secondary injury pathways, and one agent, Riluzole, has undergone Phase-III investigation in DCM. Although it did not show efficacy on the primary outcome modified Japanese Orthopaedic Association scale, it showed promising results in pain reduction. Regenerative approaches are in the early stage, with one agent, Ibudilast, currently in a phase-III investigation. Neuromodulation approaches aim to therapeutically alter the state of spinal cord excitation by electrical stimulation with a variety of approaches. Case studies using electrical neuromuscular and spinal cord stimulation have shown positive therapeutic utility. CONCLUSION There is limited research into interventions in the 3 broad areas of neuroprotection, neuroregeneration, and neuromodulation for DCM. Contemporary and novel therapies for DCM are now a top 10 priority, and whilst research in these areas is limited in DCM, it is hoped that this review will encourage research into this priority.
Collapse
Affiliation(s)
- Aref-Ali Gharooni
- Neurosurgery Unit, Department of Clinical Neuroscience, University of Cambridge, UK
| | - Brian K. Kwon
- Vancouver Spine Surgery Institute, Department of Orthopedics, The University of British Columbia, Vancouver, BC, Canada
| | - Michael G. Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Timothy F. Boerger
- Department of Neurosurgery, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - Ricardo Rodrigues-Pinto
- Spinal Unit (UVM), Department of Orthopaedics, Centro Hospitalar Universitário do Porto - Hospital de Santo António, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Paul Aarne Koljonen
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shekar N. Kurpad
- Department of Neurosurgery, Medical College of Wisconsin, Wauwatosa, WI, USA
| | - James S. Harrop
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bizhan Aarabi
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vafa Rahimi-Movaghar
- Department of Neurosurgery, Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jefferson R. Wilson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Benjamin M. Davies
- Neurosurgery Unit, Department of Clinical Neuroscience, University of Cambridge, UK
| | - Mark R. N. Kotter
- Neurosurgery Unit, Department of Clinical Neuroscience, University of Cambridge, UK
| | - James D. Guest
- Department of Neurosurgery and The Miami Project to Cure Paralysis, The Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
75
|
Walsh CM, Wychowaniec JK, Brougham DF, Dooley D. Functional hydrogels as therapeutic tools for spinal cord injury: New perspectives on immunopharmacological interventions. Pharmacol Ther 2021; 234:108043. [PMID: 34813862 DOI: 10.1016/j.pharmthera.2021.108043] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) is a complex medical and psychological challenge for which there is no curative therapy currently available. Despite major progress in pharmacological and surgical approaches, clinical trials for SCI patients have been uniformly disappointing thus far as there are many practical and biological issues yet to be resolved. Neuroinflammation is a critical event of the secondary injury phase after SCI, and recent research strategies have focused on modulating the immune response after injury to provide a more favorable recovery environment. Biomaterials can serve this purpose by providing physical and trophic support to the injured spinal cord after SCI. Of all potential biomaterials, functional hydrogels are emerging as a key component in novel treatment strategies for SCI, including controlled and localized delivery of immunomodulatory therapies to drive polarization of immune cells towards a pro-regenerative phenotype. Here, we extensively review recent developments in the use of functional hydrogels as immunomodulatory therapies for SCI. We briefly describe physicochemical properties of hydrogels and demonstrate how advanced fabrication methods lead to the required heterogeneity and hierarchical arrangements that increasingly mimic complex spinal cord tissue. We then summarize potential SCI therapeutic modalities including: (i) hydrogels alone; (ii) hydrogels as cellular or (iii) bioactive molecule delivery vehicles, and; (iv) combinatorial approaches. By linking the structural properties of hydrogels to their functions in treatment with particular focus on immunopharmacological stimuli, this may accelerate further development of functional hydrogels for SCI, and indeed next-generation central nervous system regenerative therapies.
Collapse
Affiliation(s)
- Ciara M Walsh
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jacek K Wychowaniec
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland; AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dermot F Brougham
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
76
|
Rathnam C, Yang L, Castro-Pedrido S, Luo J, Cai L, Lee KB. Hybrid SMART spheroids to enhance stem cell therapy for CNS injuries. SCIENCE ADVANCES 2021; 7:eabj2281. [PMID: 34586845 PMCID: PMC8480929 DOI: 10.1126/sciadv.abj2281] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Although stem cell therapy holds enormous potential for treating debilitating injuries and diseases in the central nervous system, low survival and inefficient differentiation have restricted its clinical applications. Recently, 3D cell culture methods, such as stem cell–based spheroids and organoids, have demonstrated advantages by incorporating tissue-mimetic 3D cell-cell interactions. However, a lack of drug and nutrient diffusion, insufficient cell-matrix interactions, and tedious fabrication procedures have compromised their therapeutic effects in vivo. To address these issues, we developed a biodegradable nanomaterial-templated 3D cell assembly method that enables the formation of hybrid stem cell spheroids with deep drug delivery capabilities and homogeneous incorporation of 3D cell-matrix interactions. Hence, high survival rates, controlled differentiation, and functional recovery were demonstrated in a spinal cord injury animal model. Overall, our hybrid stem cell spheroids represent a substantial development of material-facilitated 3D cell culture systems and can pave the way for stem cell–based treatment of CNS injuries.
Collapse
Affiliation(s)
- Christopher Rathnam
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sofia Castro-Pedrido
- Department of Biomedical Engineering Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jeffrey Luo
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Li Cai
- Department of Biomedical Engineering Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
77
|
Pinho TS, Cunha CB, Lanceros-Méndez S, Salgado AJ. Electroactive Smart Materials for Neural Tissue Regeneration. ACS APPLIED BIO MATERIALS 2021; 4:6604-6618. [PMID: 35006964 DOI: 10.1021/acsabm.1c00567] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Repair in the human nervous system is a complex and intertwined process that offers significant challenges to its study and comprehension. Taking advantage of the progress in fields such as tissue engineering and regenerative medicine, the scientific community has witnessed a strong increase of biomaterial-based approaches for neural tissue regenerative therapies. Electroactive materials, increasingly being used as sensors and actuators, also find application in neurosciences due to their ability to deliver electrical signals to the cells and tissues. The use of electrical signals for repairing impaired neural tissue therefore presents an interesting and innovative approach to bridge the gap between fundamental research and clinical applications in the next few years. In this review, first a general overview of electroactive materials, their historical origin, and characteristics are presented. Then a comprehensive view of the applications of electroactive smart materials for neural tissue regeneration is presented, with particular focus on the context of spinal cord injury and brain repair. Finally, the major challenges of the field are discussed and the main challenges for the near future presented. Overall, it is concluded that electroactive smart materials play an ever-increasing role in neural tissue regeneration, appearing as potentially valuable biomaterials for regenerative purposes.
Collapse
Affiliation(s)
- Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal.,Stemmatters, Biotecnologia e Medicina Regenerativa SA, 4805-017 Guimarães, Portugal
| | - Cristiana B Cunha
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, 4805-017 Guimarães, Portugal
| | - Senentxu Lanceros-Méndez
- Center of Physics, University of Minho, 4710-058 Braga, Portugal.,BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain.,Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
| |
Collapse
|
78
|
Zhang B, Hu L, Zhang J, Wu H, Li W, Gou L, Liu H. Insulin growth factor-1 enhances proliferation and inhibits apoptosis of neural progenitor cells by phosphorylation of Akt/mTOR/p70S6K molecules and triggering intrinsic apoptosis signaling pathway. Cell Tissue Bank 2021; 23:459-472. [PMID: 34494222 DOI: 10.1007/s10561-021-09956-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/02/2021] [Indexed: 02/05/2023]
Abstract
Neural progenitor cells (NPCs) transplantation is known as a potential strategy for treating spinal cord injury (SCI). This study aimed to investigate effects of insulin growth factor-1 (IGF-I) on NPCs proliferation and clarify associated mechanisms. NPCs isolated from T8-T10 segmental spinal cord tissues of rats were cultured and identification. Then, lentivirus packing plasmids containing IGF-I was constructed and used for NPCs infection. Cell proliferation was evaluated by detecting 5-Bromodeoxyuridine (BrdU) expression in NPCs, cell differentiation was detected using double-labeling immunofluorescence staining while cell apoptosis was detected using TUNEL assay. In addition, the signal expression of Akt/mTOR/p70S6K in NPCs cells were investigated using immunofluorescence staining and western blot assay. The experimental group was defined as pCMV-IGF-I group, while the negative control group was defined as pCMV-LacZ group. Cells infected with pCMV-IGF-I lentivirus followed by addition of 100 mg/ml rapamycin were defined as pCMV-IGF-I + Rapa group. NPCs were successfully isolated, identified and cultured. IGF-I overexpression significantly inhibited cell apoptosis and enhanced cell migration. Akt/mTOR/ p70S6K signaling cascade was proved to be present in NPCs, IGF-I overexpression significantly activated Akt/mTOR/p70S6K signaling cascade, while rapamycin addition inhibited its expression. Also, the activated Akt/mTOR/p70S6K signal cascade induced by IGF-I significantly enhanced BrdU expression and inhibited cell apoptosis, and promoted the differentiation of NPC into the neuronal system. However, the rapamycin addition inhibited the cell response induced by IGF-I overexpression. IGF-I overexpression could enhance cell proliferation, inhibit cell apoptosis and promote their differentiation into neuronal systems by activating Akt/mTOR/p70S6K signaling cascade in vitro, indicating that the Akt/mTOR/p70S6K signaling cascade may be the potentially mechanism for the endogenous repair and remodeling of spinal cord after injury.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Orthopaedic Surgery, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Lingyun Hu
- Department of Orthopaedic Surgery, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China.,Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jianying Zhang
- Department of Radiology, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Hui Wu
- Department of Orthopaedic Surgery, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Wei Li
- Department of Orthopaedic Surgery, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Lin Gou
- Department of Orthopaedic Surgery, Nanchong Central Hospital, the Second Clinical Medical College of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Hao Liu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
79
|
Wang J, Li Y, Xu T, Zhao J, Yuan C, Wen B. Reconstructing Nanohydroxyapatite Prosthesis Based on CT-Scanning Data and Its Application in Spinal Injury. J Biomed Nanotechnol 2021; 17:1745-1753. [PMID: 34688319 DOI: 10.1166/jbn.2021.3143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study investigated the nanohydroxyapatite (nHA) prosthesis application effect based on CT-scanning data in spinal injury. This study chose 26 spinal injury patients treated in our hospital from September 2017 to September 2018, who were randomly divided into two groups. nHA prosthesis based on CT-scanning data was implanted in the nHA group, whereas titanium mesh was implanted in the titanium mesh group. Consequently, osteoblasts were cultured to test the biological activity of nHA and titanium alloy. In cell tests, we found osteoblasts could better adhere to nHA, and proliferation and activity were higher when planted on nHA material. After surgical treatment, all patients' spinal symptoms (VAS score, JOA score, and Cobb angle) had improved and did not cause obvious inflammatory foreign body reactions. During a two-year follow-up, the fusion time and support settlement in the nHA group was lower, and the vertebral fusion rate and ASIA score were higher than those in the titanium mesh group. Thus, CT-scanning data could further improve the vertebral fusion rate in the nHA group. Consequentially, nHA prosthesis based on CT-scanning data is a better choice for spinal injury therapy.
Collapse
Affiliation(s)
- Jian Wang
- Departments of Radiology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi, PR China
| | - Ying Li
- Departments of Radiology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi, PR China
| | - Ting Xu
- Departments of Radiology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi, PR China
| | - Jie Zhao
- Departments of Radiology, Shanxi Provincial People's Hospital, Taiyuan 030012, Shanxi, PR China
| | - Cuihua Yuan
- Department of Orthopaedics, Affiliated Mindong Hospital of Fujian Medical University, Fuan 355000, Fujian, PR China
| | - Baojun Wen
- Department of Orthopaedics, Affiliated Mindong Hospital of Fujian Medical University, Fuan 355000, Fujian, PR China
| |
Collapse
|
80
|
Lentiviral Vectors Delivered with Biomaterials as Therapeutics for Spinal Cord Injury. Cells 2021; 10:cells10082102. [PMID: 34440872 PMCID: PMC8394044 DOI: 10.3390/cells10082102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating trauma that can cause permanent disability, life-long chronic issues for sufferers and is a big socioeconomic burden. Regenerative medicine aims to overcome injury caused deficits and restore function after SCI through gene therapy and tissue engineering approaches. SCI has a multifaceted pathophysiology. Due to this, producing therapies that target multiple different cellular and molecular mechanisms might prove to be a superior approach in attempts at regeneration. Both biomaterials and nucleic acid delivery via lentiviral vectors (LVs) have proven to promote repair and restoration of function post SCI in animal models. Studies indicate that a combination of biomaterials and LVs is more effective than either approach alone. This review presents studies supporting the use of LVs and LVs delivered with biomaterials in therapies for SCI and summarises methods to combine LVs with biomaterials for SCI treatment. By summarising this knowledge this review aims to demonstrate how LV delivery with biomaterials can augment/compliment both LV and biomaterial therapeutic effects in SCI.
Collapse
|
81
|
Jahandideh A, Noori H, Rahimi B, Hamblin MR, Behroozi Z, Ramezani M, Ramezani F. Alginate scaffolds improve functional recovery after spinal cord injury. Eur J Trauma Emerg Surg 2021; 48:1711-1721. [PMID: 34363487 DOI: 10.1007/s00068-021-01760-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE In this systematic review and meta-analysis, the use of alginate for the repair of the damaged spinal cord was investigated. METHODS After an extensive search of databases including MEDLINE, SCOPUS, EMBASE and Web of Science, an initial screening was performed based on inclusion and exclusion criteria. The full text of related articles was reviewed and data mining was performed. Data were analyzed by calculating the mean of ratios between treated and untreated groups using STATA software. Subgroup analysis was also performed due to heterogeneity. Articles were subjected to quality control and PRISMA guidelines were followed. RESULTS Twelve studies and 17 experiments were included in the study. After SCI, alginate hydrogel had a moderate effect on motor function recovery (SMD = 0.64; 95% CI 0.28-1.00; p < 0.0001) and alginate scaffolds loaded with drugs, growth factors, or cells on the SCI group compared with untreated SCI animals showed has a strong effect in the treatment of SCI (SMD = 2.82; 95% CI 1.49-4.145; p < 0.0001). Treatment with drug/cell in combination with alginate was more strongly significant compared to the groups treated with drug/cell alone (SMD = 4.55; 95% CI 1.42-7.69; p < 0.0001). Alginate alone or in combination therapy when used as an implant, had a more significant effect than injection. CONCLUSION These findings suggest that alginate is an efficient scaffold for functional recovery and even a much better scaffold for drug/cell delivery after SCI.
Collapse
Affiliation(s)
- Atefeh Jahandideh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Noori
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Rahimi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Zahra Behroozi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
82
|
Use of electroconductive biomaterials for engineering tissues by 3D printing and 3D bioprinting. Essays Biochem 2021; 65:441-466. [PMID: 34296738 DOI: 10.1042/ebc20210003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
Existing methods of engineering alternatives to restore or replace damaged or lost tissues are not satisfactory due to the lack of suitable constructs that can fit precisely, function properly and integrate into host tissues. Recently, three-dimensional (3D) bioprinting approaches have been developed to enable the fabrication of pre-programmed synthetic tissue constructs that have precise geometries and controlled cellular composition and spatial distribution. New bioinks with electroconductive properties have the potential to influence cellular fates and function for directed healing of different tissue types including bone, heart and nervous tissue with the possibility of improved outcomes. In the present paper, we review the use of electroconductive biomaterials for the engineering of tissues via 3D printing and 3D bioprinting. Despite significant advances, there remain challenges to effective tissue replacement and we address these challenges and describe new approaches to advanced tissue engineering.
Collapse
|
83
|
Suzuki H, Sakai T. Current Concepts of Stem Cell Therapy for Chronic Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms22147435. [PMID: 34299053 PMCID: PMC8308009 DOI: 10.3390/ijms22147435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in clinical trials. In addition, several more are coming down the translational pipeline. Among ongoing and completed trials are those reporting the use of mesenchymal stem cells, neural stem/progenitor cells, induced pluripotent stem cells, olfactory ensheathing cells, and Schwann cells. The advancements in stem cell technology, combined with the powerful neuroimaging modalities, can now accelerate the pathway of promising novel therapeutic strategies from bench to bedside. Various combinations of different molecular therapies have been combined with supportive scaffolds to facilitate favorable cell–material interactions. In this review, we summarized some of the most recent insights into the preclinical and clinical studies using stem cells and other supportive drugs to unlock the microenvironment in chronic SCI to treat patients with this condition. Successful future therapies will require these stem cells and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, loss of structural framework, and immunorejection.
Collapse
|
84
|
Roy A, Pathak Z, Kumar H. Strategies to neutralize RhoA/ROCK pathway after spinal cord injury. Exp Neurol 2021; 343:113794. [PMID: 34166685 DOI: 10.1016/j.expneurol.2021.113794] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/01/2021] [Accepted: 06/19/2021] [Indexed: 01/22/2023]
Abstract
Regeneration is bungled following CNS injuries, including spinal cord injury (SCI). Inherent decay of permissive conditions restricts the regrowth of the mature CNS after an injury. Hypertrophic scarring, insignificant intrinsic axon-growth activity, and axon-growth inhibitory molecules such as myelin inhibitors and scar inhibitors constitute a significant hindrance to spinal cord repair. Besides these molecules, a combined absence of various mechanisms responsible for axonal regeneration is the main reason behind the dereliction of the adult CNS to regenerate. The neutralization of specific inhibitors/proteins by stymieing antibodies or encouraging enzymatic degradation results in improved axon regeneration. Previous efforts to induce regeneration after SCI have stimulated axonal development in or near lesion sites, but not beyond them. Several pathways are responsible for the axonal growth obstruction after a CNS injury, including SCI. Herein, we summarize the axonal, glial, and intrinsic factor which impedes the regeneration. We have also discussed the methods to stabilize microtubules and through this to maintain the proper cytoskeletal dynamics of growth cone as disorganized microtubules lead to the failure of axonal regeneration. Moreover, we primarily focus on diverse inhibitors of axonal growth and molecular approaches to counteract them and their downstream intracellular signaling through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
85
|
Babu S, Albertino F, Omidinia Anarkoli A, De Laporte L. Controlling Structure with Injectable Biomaterials to Better Mimic Tissue Heterogeneity and Anisotropy. Adv Healthc Mater 2021; 10:e2002221. [PMID: 33951341 PMCID: PMC11469279 DOI: 10.1002/adhm.202002221] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/17/2021] [Indexed: 12/15/2022]
Abstract
Tissue regeneration of sensitive tissues calls for injectable scaffolds, which are minimally invasive and offer minimal damage to the native tissues. However, most of these systems are inherently isotropic and do not mimic the complex hierarchically ordered nature of the native extracellular matrices. This review focuses on the different approaches developed in the past decade to bring in some form of anisotropy to the conventional injectable tissue regenerative matrices. These approaches include introduction of macroporosity, in vivo pattering to present biomolecules in a spatially and temporally controlled manner, availability of aligned domains by means of self-assembly or oriented injectable components, and in vivo bioprinting to obtain structures with features of high resolution that resembles native tissues. Toward the end of the review, different techniques to produce building blocks for the fabrication of heterogeneous injectable scaffolds are discussed. The advantages and shortcomings of each approach are discussed in detail with ideas to improve the functionality and versatility of the building blocks.
Collapse
Affiliation(s)
- Susan Babu
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 2Aachen52074Germany
- DWI‐Leibniz Institute for Interactive MaterialsForckenbeckstrasse 50Aachen52074Germany
- Max Planck School‐Matter to Life (MtL)Jahnstrasse 29Heidelberg69120Germany
| | - Filippo Albertino
- DWI‐Leibniz Institute for Interactive MaterialsForckenbeckstrasse 50Aachen52074Germany
| | | | - Laura De Laporte
- Institute of Technical and Macromolecular Chemistry (ITMC)Polymeric BiomaterialsRWTH University AachenWorringerweg 2Aachen52074Germany
- DWI‐Leibniz Institute for Interactive MaterialsForckenbeckstrasse 50Aachen52074Germany
- Max Planck School‐Matter to Life (MtL)Jahnstrasse 29Heidelberg69120Germany
- Advanced Materials for Biomedicine (AMB)Institute of Applied Medical Engineering (AME)Center for Biohybrid Medical Systems (CMBS)University Hospital RWTH AachenForckenbeckstrasse 55Aachen52074Germany
| |
Collapse
|
86
|
One Raft to Guide Them All, and in Axon Regeneration Inhibit Them. Int J Mol Sci 2021; 22:ijms22095009. [PMID: 34066896 PMCID: PMC8125918 DOI: 10.3390/ijms22095009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
Central nervous system damage caused by traumatic injuries, iatrogenicity due to surgical interventions, stroke and neurodegenerative diseases is one of the most prevalent reasons for physical disability worldwide. During development, axons must elongate from the neuronal cell body to contact their precise target cell and establish functional connections. However, the capacity of the adult nervous system to restore its functionality after injury is limited. Given the inefficacy of the nervous system to heal and regenerate after damage, new therapies are under investigation to enhance axonal regeneration. Axon guidance cues and receptors, as well as the molecular machinery activated after nervous system damage, are organized into lipid raft microdomains, a term typically used to describe nanoscale membrane domains enriched in cholesterol and glycosphingolipids that act as signaling platforms for certain transmembrane proteins. Here, we systematically review the most recent findings that link the stability of lipid rafts and their composition with the capacity of axons to regenerate and rebuild functional neural circuits after damage.
Collapse
|
87
|
Kim KD, Lee KS, Coric D, Chang JJ, Harrop JS, Theodore N, Toselli RM. A study of probable benefit of a bioresorbable polymer scaffold for safety and neurological recovery in patients with complete thoracic spinal cord injury: 6-month results from the INSPIRE study. J Neurosurg Spine 2021; 34:808-817. [PMID: 33545674 DOI: 10.3171/2020.8.spine191507] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/17/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate whether the investigational Neuro-Spinal Scaffold (NSS), a highly porous bioresorbable polymer device, demonstrates probable benefit for safety and neurological recovery in patients with complete (AIS grade A) T2-12 spinal cord injury (SCI) when implanted ≤ 96 hours postinjury. METHODS This was a prospective, open-label, multicenter, single-arm study in patients with a visible contusion on MRI. The NSS was implanted into the epicenter of the postirrigation intramedullary spinal cord contusion cavity with the intention of providing structural support to the injured spinal cord parenchyma. The primary efficacy endpoint was the proportion of patients who had an improvement of ≥ 1 AIS grade (i.e., conversion from complete paraplegia to incomplete paraplegia) at the 6-month follow-up visit. A preset objective performance criterion established for the study was defined as an AIS grade conversion rate of ≥ 25%. Secondary endpoints included change in neurological level of injury (NLI). This analysis reports on data through 6-month follow-up assessments. RESULTS Nineteen patients underwent NSS implantation. There were 3 early withdrawals due to death, which were all determined by investigators to be unrelated to the NSS or the implantation procedure. Seven of 16 patients (43.8%) who completed the 6-month follow-up visit had conversion of neurological status (AIS grade A to grade B [n = 5] or C [n = 2]). Five patients showed improvement in NLI of 1 to 2 levels compared with preimplantation assessment, 3 patients showed no change, and 8 patients showed deterioration of 1 to 4 levels. There were no unanticipated or serious adverse device effects or serious adverse events related to the NSS or the implantation procedure as determined by investigators. CONCLUSIONS In this first-in-human study, implantation of the NSS within the spinal cord appeared to be safe in the setting of surgical decompression and stabilization for complete (AIS grade A) thoracic SCI. It was associated with a 6-month AIS grade conversion rate that exceeded historical controls. The INSPIRE study data demonstrate that the potential benefits of the NSS outweigh the risks in this patient population and support further clinical investigation in a randomized controlled trial. Clinical trial registration no.: NCT02138110 (clinicaltrials.gov).
Collapse
Affiliation(s)
- Kee D Kim
- 1Department of Neurological Surgery, UC Davis, Sacramento, California
| | - K Stuart Lee
- 2Division of Neurosurgery, Vidant Health, Greenville, North Carolina
| | - Domagoj Coric
- 3Atrium Musculoskeletal Institute, Carolina NeuroSurgery & Spine Associates, Charlotte, North Carolina
| | - Jason J Chang
- 4Department of Neurological Surgery, Oregon Health and Sciences University, Portland, Oregon
| | - James S Harrop
- 5Department of Neurological and Orthopedic Surgery, Division of Spine and Peripheral Nerve Surgery, and Delaware Valley SCI Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Nicholas Theodore
- 6Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland; and
| | | |
Collapse
|
88
|
Ciciriello AJ, Smith DR, Munsell MK, Boyd SJ, Shea LD, Dumont CM. IL-10 lentivirus-laden hydrogel tubes increase spinal progenitor survival and neuronal differentiation after spinal cord injury. Biotechnol Bioeng 2021; 118:2609-2625. [PMID: 33835500 DOI: 10.1002/bit.27781] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
A complex cellular cascade characterizes the pathophysiological response following spinal cord injury (SCI) limiting regeneration. Biomaterial and stem cell combination therapies together have shown synergistic effects, compared to the independent benefits of each intervention, and represent a promising approach towards regaining function after injury. In this study, we combine our polyethylene glycol (PEG) cell delivery platform with lentiviral-mediated overexpression of the anti-inflammatory cytokine interleukin (IL)-10 to improve mouse embryonic Day 14 (E14) spinal progenitor transplant survival. Immediately following injury in a mouse SCI hemisection model, five PEG tubes were implanted followed by direct injection into the tubes of lentivirus encoding for IL-10. Two weeks after tube implantation, mouse E14 spinal progenitors were injected directly into the integrated tubes, which served as a soft substrate for cell transplantation. Together, the tubes with the IL-10 encoding lentivirus improved E14 spinal progenitor survival, assessed at 2 weeks posttransplantation (4 weeks postinjury). On average, 8.1% of E14 spinal progenitors survived in mice receiving IL-10 lentivirus-laden tubes compared with 0.7% in mice receiving transplants without tubes, an 11.5-fold difference. Surviving E14 spinal progenitors gave rise to neurons when injected into tubes. Axon elongation and remyelination were observed, in addition to a significant increase in functional recovery in mice receiving IL-10 lentivirus-laden tubes with E14 spinal progenitor delivery compared to the injury only control by 4 weeks postinjury. All other conditions did not exhibit increased stepping until 8 or 12 weeks postinjury. This system affords increased control over the transplantation microenvironment, offering the potential to improve stem cell-mediated tissue regeneration.
Collapse
Affiliation(s)
- Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,DJTMF Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Mary K Munsell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Sydney J Boyd
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,DJTMF Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| |
Collapse
|
89
|
Lee HL, Yeum CE, Lee H, Oh J, Kim JT, Lee WJ, Ha Y, Yang YI, Kim KN. Peripheral Nerve-Derived Stem Cell Spheroids Induce Functional Recovery and Repair after Spinal Cord Injury in Rodents. Int J Mol Sci 2021; 22:ijms22084141. [PMID: 33923671 PMCID: PMC8072978 DOI: 10.3390/ijms22084141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy is one of the most promising candidate treatments for spinal cord injury. Research has shown optimistic results for this therapy, but clinical limitations remain, including poor viability, engraftment, and differentiation. Here, we isolated novel peripheral nerve-derived stem cells (PNSCs) from adult peripheral nerves with similar characteristics to neural-crest stem cells. These PNSCs expressed neural-crest specific markers and showed multilineage differentiation potential into Schwann cells, neuroglia, neurons, and mesodermal cells. In addition, PNSCs showed therapeutic potential by releasing the neurotrophic factors, including glial cell-line-derived neurotrophic factor, insulin-like growth factor, nerve growth factor, and neurotrophin-3. PNSC abilities were also enhanced by their development into spheroids which secreted neurotrophic factors several times more than non-spheroid PNSCs and expressed several types of extra cellular matrix. These features suggest that the potential for these PNSC spheroids can overcome their limitations. In an animal spinal cord injury (SCI) model, these PNSC spheroids induced functional recovery and neuronal regeneration. These PNSC spheroids also reduced the neuropathic pain which accompanies SCI after remyelination. These PNSC spheroids may represent a new therapeutic approach for patients suffering from SCI.
Collapse
Affiliation(s)
- Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Chung-Eun Yeum
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - HyeYeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jinsoo Oh
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
| | - Jong-Tae Kim
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Won-Jin Lee
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Korea
| | - Young-Il Yang
- Paik Inje Memorial Institute for Clinical Research, Inje University College of Medicine, Busan 47392, Korea; (C.-E.Y.); (J.-T.K.); (W.-J.L.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| | - Keung-Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul 03722, Korea; (H.-L.L.); (H.L.); (J.O.); (Y.H.)
- Correspondence: (Y.-I.Y.); (K.-N.K.)
| |
Collapse
|
90
|
Ehsanipour A, Sathialingam M, Rad LM, de Rutte J, Bierman RD, Liang J, Xiao W, Di Carlo D, Seidlits SK. Injectable, macroporous scaffolds for delivery of therapeutic genes to the injured spinal cord. APL Bioeng 2021; 5:016104. [PMID: 33728392 PMCID: PMC7946441 DOI: 10.1063/5.0035291] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Biomaterials are being developed as therapeutics for spinal cord injury (SCI) that can stabilize and bridge acute lesions and mediate the delivery of transgenes, providing a localized and sustained reservoir of regenerative factors. For clinical use, direct injection of biomaterial scaffolds is preferred to enable conformation to unique lesions and minimize tissue damage. While an interconnected network of cell-sized macropores is necessary for rapid host cell infiltration into-and thus integration of host tissue with-implanted scaffolds, injectable biomaterials have generally suffered from a lack of control over the macrostructure. As genetic vectors have short lifetimes in vivo, rapid host cell infiltration into scaffolds is a prerequisite for efficient biomaterial-mediated delivery of transgenes. We present scaffolds that can be injected and assembled in situ from hyaluronic acid (HA)-based, spherical microparticles to form scaffolds with a network of macropores (∼10 μm). The results demonstrate that addition of regularly sized macropores to traditional hydrogel scaffolds, which have nanopores (∼10 nm), significantly increases the expression of locally delivered transgene to the spinal cord after a thoracic injury. Maximal cell and axon infiltration into scaffolds was observed in scaffolds with more regularly sized macropores. The delivery of lentiviral vectors encoding the brain-derived neurotrophic factor (BDNF), but not neurotrophin-3, from these scaffolds further increased total numbers and myelination of infiltrating axons. Modest improvements to the hindlimb function were observed with BDNF delivery. The results demonstrate the utility of macroporous and injectable HA scaffolds as a platform for localized gene therapies after SCI.
Collapse
Affiliation(s)
- Arshia Ehsanipour
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Mayilone Sathialingam
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Laila M Rad
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Joseph de Rutte
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Rebecca D Bierman
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Jesse Liang
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Weikun Xiao
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
91
|
Zhang H, Piao M, Guo M, Meng L, Yu H. MicroRNA-211-5p attenuates spinal cord injury via targeting of activating transcription factor 6. Tissue Cell 2021; 68:101459. [PMID: 33238217 DOI: 10.1016/j.tice.2020.101459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
The recovery of spinal cord injury (SCI) involves multiple factors, of which miRNAs take an important part. In this study, we evaluated the function of microRNA-211-5p (miR-211-5p) on SCI in a rat model. SCI model was established using modified Allen's weight-drop method and Basso-Bcattie-Bresnahan score was applied to assess the locomotor function. MiR-211-5p agomir was utilized to increase miR-211-5p expression and endoplasmic reticulum (ER) stress inhibitor, 4-PBA (4-phenylbutyric acid), was utilized to suppress ER stress. Neuron apoptosis and the expressions of miR-211-5p, activating transcription factor 6 (ATF6), apoptosis-related proteins, pro-inflammatory cytokines and endoplasmic reticulum stress-related proteins were detected. Dual luciferase reporter gene assay was performed to verify the binding between miR-211-5p and ATF6. The results showed that miR-211-5p directly targeted ATF6. MiR-211-5p was down-regulated and ATF6 was up-regulated in SCI rats. Both interferences with miR-211-5p agomir and 4-PBA effectively attenuated neuron apoptosis and reversed the expressions of apoptosis, inflammation and endoplasmic reticulum stress-related molecules post SCI in rats. These findings demonstrated that miR-211-5p could effectively alleviate SCI-induced neuron apoptosis and inflammation via directly targeting ATF-6 and regulating ER stress.
Collapse
Affiliation(s)
- Haocong Zhang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110016, China
| | - Meihui Piao
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110016, China
| | - Mingming Guo
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110016, China
| | - Lingzhi Meng
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110016, China
| | - Hailong Yu
- Department of Orthopaedics, The General Hospital of Northern Theater Command, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
92
|
Wei L, Cai G, Jiang L, Gao L, Yang Z, Zhang W. Identification of key pathways and RNAs associated with skeletal muscle atrophy after spinal cord injury. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2021; 21:550-559. [PMID: 34854395 PMCID: PMC8672411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This study was performed to investigate the potential key molecules involved in the progression of skeletal muscle atrophy after SCI. METHODS Based on GSE21497 dataset, the DEmRNAs and DElncRNAs were screened after differentially expressed analysis. Then the enrichment analyses were performed on DEmRNAs. Then the PPI network and ceRNA network were constructed. Finally, the DGIdb was utilized to predict drug-gene interactions. RESULTS A total of 412 DEmRNAs and 21 DElncRNAs were obtained. The DEmRNAs were significantly enriched in MAPK signaling pathway and FoxO signaling pathway. In addition, UBE2D1, JUN, and FBXO32 had higher node degrees in PPI network, and the top 20 genes with high degree were significantly enriched in FoxO signaling pathway and Endometrial cancer. Moreover, FOXO3 was regulated by hsa-miR-1207-5p and hsa-miR-1207-5p was regulated by lncRNA RP11-253E3.3 in ceRNA network. Finally, 37 drug-gene interactions were obtained based on the 26 genes in ceRNA network. CONCLUSION UBE2D1, JUN, and FBXO32 are likely to be related to the progression of skeletal muscle atrophy after SCI, and activating of MAPK signaling pathway, Endometrial cancer and FoxO signaling pathway may induce skeletal muscle inflammation, apoptosis, autophagy and atrophy after SCI. Moreover, RP11-253E3.3-hsa-miR-1207-5p-FOXO3 axis may be a promising therapeutic target for skeletal muscle atrophy after SCI.
Collapse
Affiliation(s)
- Li Wei
- Department of Rehabilitation Medicine, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, Shanghai, China
| | - Guoying Cai
- Preventive Treatment Department, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, Shanghai, China
| | - Lian Jiang
- Department of Rehabilitation Medicine, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, Shanghai, China
| | - Linhui Gao
- Department of Rehabilitation Medicine, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, Shanghai, China
| | - Zehui Yang
- Department of Rehabilitation Medicine, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, Shanghai, China
| | - Wei Zhang
- Department of Rehabilitation Medicine, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, Shanghai, China,Corresponding author: Wei Zhang, Department of Rehabilitation Medicine, Minhang District Integrated Hospitals of Traditional Chinese and Western Medicine, No.155 Jianchuan Road, Minhang District, Shanghai, 200241, China E-mail:
| |
Collapse
|
93
|
Cliver RN, Ayers B, Brady A, Firestein BL, Vazquez M. Cerebrospinal fluid replacement solutions promote neuroglia migratory behaviors and spinal explant outgrowth in microfluidic culture. J Tissue Eng Regen Med 2020; 15:176-188. [PMID: 33274811 DOI: 10.1002/term.3164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/23/2022]
Abstract
Disorders of the nervous system (NS) impact millions of adults, worldwide, as a consequence of traumatic injury, genetic illness, or chronic health conditions. Contemporary studies have begun to incorporate neuroglia into emerging NS therapies to harness the regenerative potential of glial-mediated synapses in the brain and spinal cord. However, the role of cerebrospinal fluid (CSF) that surrounds neuroglia and interfaces with their associated synapses remains only partially explored. The flow of CSF within subarachnoid spaces (SAS) circulates essential polypeptides, metabolites, and growth factors that directly impact neural response and recovery via signaling with healthy glia. Despite the availability of artificial CSF solutions used in neurosurgery and NS treatments, tissue engineering projects continue to use cell culture media, such as Neurobasal (NB) and Dulbecco's Modified Eagle Medium (DMEM), for development and characterization of many transplantable cells, matrixes, and integrated cellular systems. The current study examined in vitro behaviors of glial Schwann cells (ShC) and spinal cord explants (SCE) within a CSF replacement solution, Elliott's B Solution (EBS), used widely in the treatment of NS disorders. Our tests used EBS to create defined chemical microenvironments of extracellular factors within a glial line (gLL) microfluidic device, previously described by our group. The gLL is comparable in scale to the in vivo SAS that envelopes endogenous CSF and enables molecular transport via mechanisms of convective diffusion. Our results illustrate that EBS solutions facilitate ShC survival, morphology, and proliferation similar to those measured in traditional DMEM, and additionally support glial chemotactic behaviors in response to brain-derived growth factor (BDNF). Our data indicates that ShC undergo significant chemotaxis toward high and low concentration gradients of BDNF with statistical differences between gradients formed within diluents of EBS and DMEM solutions. Moreover, SCE cultured with EBS solutions facilitated measurement of neurite explant extension commensurate with reported in vivo measurements. This data highlights the translational significance and advantages of incorporating CSF replacement fluids to interrogate cellular behaviors and advance regenerative NS therapies.
Collapse
Affiliation(s)
- Richard N Cliver
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Brian Ayers
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Alyssa Brady
- Department of Physics, Salisbury University, Salisbury, Maryland, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
94
|
Zimmermann R, Vieira Alves Y, Sperling LE, Pranke P. Nanotechnology for the Treatment of Spinal Cord Injury. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:353-365. [PMID: 33135599 DOI: 10.1089/ten.teb.2020.0188] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) affects the central nervous system (CNS) and there is currently no treatment with the potential for rehabilitation. Although several clinical treatments have been developed, they are still at an early stage and have not shown success in repairing the broken fiber, which prevents cellular regeneration and integral restoration of motor and sensory functions. Considering the importance of nanotechnology and tissue engineering for neural tissue injuries, this review focuses on the latest advances in nanotechnology for SCI treatment and tissue repair. The PubMed database was used for the bibliographic survey. Initial research using the following keywords "tissue engineering and spinal cord injury" revealed 970 articles published in the last 10 years. The articles were further analyzed, excluding those not related to SCI or with results that did not pertain to the field of interest, including the reviews. It was observed that a total of 811 original articles used the quoted keywords. When the word "treatment" was added, 662 articles were found and among them, 529 were original ones. Finally, when the keywords "Nanotechnology and spinal cord injury" were used, 102 articles were found, 65 being original articles. A search concerning the biomaterials used for SCI found 700 articles with 589 original articles. A total of 107 articles were included in the discussion of this review and some are used for the theoretical framework. Recent progress in nanotechnology and tissue engineering has shown promise for repairing CNS damage. A variety of in vivo animal testing for SCI has been used with or without cells and some of these in vivo studies have shown successful results. However, there is no translation to humans using nanotechnology for SCI treatment, although there is one ongoing trial that employs a tissue engineering approach, among other technologies. The first human surgical scaffold implantation will elucidate the possibility of this use for further clinical trials. This review concludes that even though tissue engineering and nanotechnology are being investigated as a possibility for SCI treatment, tests with humans are still in the theoretical stage. Impact statement Thousands of people are affected by spinal cord injury (SCI) per year in the world. This type of lesion is one of the most severe conditions that can affect humans and usually causes permanent loss of strength, sensitivity, and motor function below the injury site. This article reviews studies on the PubMed database, assessing the publications on SCI in the study field of tissue engineering, focusing on the use of nanotechnology for the treatment of SCI. The review makes an evaluation of the biomaterials used for the treatment of this condition and the techniques applied for the production of nanostructured biomaterials.
Collapse
Affiliation(s)
- Rafaela Zimmermann
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Yuri Vieira Alves
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Laura E Sperling
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Health School, Faculty of Medicine, UNISINOS, São Leopoldo, Brazil
| | - Patricia Pranke
- Hematology and Stem Cell Laboratory, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Stem Cell Research Institute, Porto Alegre, Brazil
| |
Collapse
|
95
|
Spinal Cord Injury: Pathophysiology, Multimolecular Interactions, and Underlying Recovery Mechanisms. Int J Mol Sci 2020; 21:ijms21207533. [PMID: 33066029 PMCID: PMC7589539 DOI: 10.3390/ijms21207533] [Citation(s) in RCA: 663] [Impact Index Per Article: 132.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) is a destructive neurological and pathological state that causes major motor, sensory and autonomic dysfunctions. Its pathophysiology comprises acute and chronic phases and incorporates a cascade of destructive events such as ischemia, oxidative stress, inflammatory events, apoptotic pathways and locomotor dysfunctions. Many therapeutic strategies have been proposed to overcome neurodegenerative events and reduce secondary neuronal damage. Efforts have also been devoted in developing neuroprotective and neuro-regenerative therapies that promote neuronal recovery and outcome. Although varying degrees of success have been achieved, curative accomplishment is still elusive probably due to the complex healing and protective mechanisms involved. Thus, current understanding in this area must be assessed to formulate appropriate treatment modalities to improve SCI recovery. This review aims to promote the understanding of SCI pathophysiology, interrelated or interlinked multimolecular interactions and various methods of neuronal recovery i.e., neuroprotective, immunomodulatory and neuro-regenerative pathways and relevant approaches.
Collapse
|
96
|
Yang T, Xing L, Yu W, Cai Y, Cui S, Chen G. Astrocytic reprogramming combined with rehabilitation strategy improves recovery from spinal cord injury. FASEB J 2020; 34:15504-15515. [PMID: 32975845 DOI: 10.1096/fj.202001657rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 01/28/2023]
Abstract
After spinal cord injury (SCI), the irreversible loss of neurons and the dense glial scar are two of the leading causes of axon regeneration failure. The adult mammalian spinal cord lacks the ability to spontaneously produce new neurons, making it a key challenge to provide new neurons for spinal cord regeneration. Additionally, the dual role of the glial scar (both inhibitory and protective) makes it difficult to manipulate it for therapeutic purposes. In this study, using a single transcription factor Sry-related HMG-box 2 (Sox2) delivered by adeno-associated virus (AAV), we reprogrammed some of the astrocytes targeted by the viral vectors in the glial scar into neurons in a severe SCI model. We show that this astrocytic reprogramming alone can propel axon regeneration by not only replenishing the lost neurons, but also moderately reducing the density of the glial scar without interrupting its integrity. Beyond that, astrocytic reprogramming can significantly improve functional recovery when combined with running wheel rehabilitation, which provides use-dependent plasticity. These findings may provide us with a new idea for how to manipulate the glial scar and a promising therapeutic strategy that combines biological intervention with a rehabilitation strategy.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.,Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Weiwei Yu
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yunyun Cai
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shusen Cui
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Gang Chen
- Department of Tissue and Embryology, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
97
|
Animal Models of Cerebral Changes Secondary to Spinal Cord Injury. World Neurosurg 2020; 145:244-250. [PMID: 32980567 DOI: 10.1016/j.wneu.2020.09.103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 11/20/2022]
Abstract
Spinal cord injuries (SCIs) are difficult to treat. The first animal SCI model (featuring the dropping of a weight) was established by Allen in 1911, and other animal models have been developed since then. Most animal studies have focused only on the molecular features of SCIs, which remain disputed. Recently, it has become clear that SCI may trigger mental and cognitive disorders, however, and brain changes secondary to SCI are under investigation. No consensus on an optimal animal model for cerebral research has emerged. We discuss the appropriate SCI models for studying secondary brain changes.
Collapse
|
98
|
Bartlett RD, Burley S, Ip M, Phillips JB, Choi D. Cell Therapies for Spinal Cord Injury: Trends and Challenges of Current Clinical Trials. Neurosurgery 2020; 87:E456-E472. [DOI: 10.1093/neuros/nyaa149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 03/11/2020] [Indexed: 12/22/2022] Open
Abstract
Abstract
Cell therapies have the potential to revolutionize the treatment of spinal cord injury. Basic research has progressed significantly in recent years, with a plethora of cell types now reaching early-phase human clinical trials, offering new strategies to repair the spinal cord. However, despite initial enthusiasm for preclinical and early-phase clinical trials, there has been a notable hiatus in the translation of cell therapies to routine clinical practice. Here, we review cell therapies that have reached clinical trials for spinal cord injury, providing a snapshot of all registered human trials and a summary of all published studies. Of registered trials, the majority have used autologous cells and approximately a third have been government funded, a third industry sponsored, and a third funded by university or healthcare systems. A total of 37 cell therapy trials have been published, primarily using stem cells, although a smaller number have used Schwann cells or olfactory ensheathing cells. Significant challenges remain for cell therapy trials in this area, including achieving stringent regulatory standards, ensuring appropriately powered efficacy trials, and establishing sustainable long-term funding. However, cell therapies hold great promise for human spinal cord repair and future trials must continue to capitalize on the exciting developments emerging from preclinical studies.
Collapse
Affiliation(s)
- Richard D Bartlett
- Centre for Nerve Engineering, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
| | - Sarah Burley
- Centre for Nerve Engineering, University College London, London, United Kingdom
| | - Mina Ip
- Centre for Nerve Engineering, University College London, London, United Kingdom
| | - James B Phillips
- Centre for Nerve Engineering, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - David Choi
- Centre for Nerve Engineering, University College London, London, United Kingdom
- Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
99
|
Lee K, Xue Y, Lee J, Kim HJ, Liu Y, Tebon P, Sarikhani E, Sun W, Zhang S, Haghniaz R, Çelebi-Saltik B, Zhou X, Ostrovidov S, Ahadian S, Ashammakhi N, Dokmeci MR, Khademhosseini A. A Patch of Detachable Hybrid Microneedle Depot for Localized Delivery of Mesenchymal Stem Cells in Regeneration Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2000086. [PMID: 33071712 PMCID: PMC7567343 DOI: 10.1002/adfm.202000086] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/20/2020] [Indexed: 05/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have been widely used for regenerative therapy. In most current clinical applications, MSCs are delivered by injection but face significant issues with cell viability and penetration into the target tissue due to a limited migration capacity. Some therapies have attempted to improve MSC stability by their encapsulation within biomaterials; however, these treatments still require an enormous number of cells to achieve therapeutic efficacy due to low efficiency. Additionally, while local injection allows for targeted delivery, injections with conventional syringes are highly invasive. Due to the challenges associated with stem cell delivery, a local and minimally invasive approach with high efficiency and improved cell viability is highly desired. In this study, we present a detachable hybrid microneedle depot (d-HMND) for cell delivery. Our system consists of an array of microneedles with an outer poly(lactic-co-glycolic) acid (PLGA) shell and an internal gelatin methacryloyl (GelMA)-MSC mixture (GMM). The GMM was characterized and optimized for cell viability and mechanical strength of the d-HMND required to penetrate mouse skin tissue was also determined. MSC viability and function within the d-HMND was characterized in vitro and the regenerative efficacy of the d-HMND was demonstrated in vivo using a mouse skin wound model.
Collapse
Affiliation(s)
- KangJu Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yumeng Xue
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Junmin Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Han-Jun Kim
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yaowen Liu
- College of Food Science, Sichuan Agricultural University, Yaan, 625014, China
| | - Peyton Tebon
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Einollah Sarikhani
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wujin Sun
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shiming Zhang
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Reihaneh Haghniaz
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Betül Çelebi-Saltik
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Xingwu Zhou
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Serge Ostrovidov
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samad Ahadian
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nureddin Ashammakhi
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mehmet R. Dokmeci
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ali Khademhosseini
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
100
|
Papadimitriou L, Manganas P, Ranella A, Stratakis E. Biofabrication for neural tissue engineering applications. Mater Today Bio 2020; 6:100043. [PMID: 32190832 PMCID: PMC7068131 DOI: 10.1016/j.mtbio.2020.100043] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/28/2022] Open
Abstract
Unlike other tissue types, the nervous tissue extends to a wide and complex environment that provides a plurality of different biochemical and topological stimuli, which in turn defines the advanced functions of that tissue. As a consequence of such complexity, the traditional transplantation therapeutic methods are quite ineffective; therefore, the restoration of peripheral and central nervous system injuries has been a continuous scientific challenge. Tissue engineering and regenerative medicine in the nervous system have provided new alternative medical approaches. These methods use external biomaterial supports, known as scaffolds, to create platforms for the cells to migrate to the injury site and repair the tissue. The challenge in neural tissue engineering (NTE) remains the fabrication of scaffolds with precisely controlled, tunable topography, biochemical cues, and surface energy, capable of directing and controlling the function of neuronal cells toward the recovery from neurological disorders and injuries. At the same time, it has been shown that NTE provides the potential to model neurological diseases in vitro, mainly via lab-on-a-chip systems, especially in cases for which it is difficult to obtain suitable animal models. As a consequence of the intense research activity in the field, a variety of synthetic approaches and 3D fabrication methods have been developed for the fabrication of NTE scaffolds, including soft lithography and self-assembly, as well as subtractive (top-down) and additive (bottom-up) manufacturing. This article aims at reviewing the existing research effort in the rapidly growing field related to the development of biomaterial scaffolds and lab-on-a-chip systems for NTE applications. Besides presenting recent advances achieved by NTE strategies, this work also delineates existing limitations and highlights emerging possibilities and future prospects in this field.
Collapse
Affiliation(s)
- L. Papadimitriou
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - P. Manganas
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - A. Ranella
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - E. Stratakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
- Physics Department, University of Crete, Heraklion, 71003, Crete, Greece
| |
Collapse
|