1
|
Moosavi SG, Rahiman N, Jaafari MR, Arabi L. Lipid nanoparticle (LNP) mediated mRNA delivery in neurodegenerative diseases. J Control Release 2025; 381:113641. [PMID: 40120689 DOI: 10.1016/j.jconrel.2025.113641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Neurodegenerative diseases (NDD) are characterized by the progressive loss of neurons and the impairment of cellular functions. Messenger RNA (mRNA) has emerged as a promising therapy for treating NDD, as it can encode missing or dysfunctional proteins and anti-inflammatory cytokines or neuroprotective proteins to halt the progression of these diseases. However, effective mRNA delivery to the central nervous system (CNS) remains a significant challenge due to the limited penetration of the blood-brain barrier (BBB). Lipid nanoparticles (LNPs) offer an efficient solution by encapsulating and protecting mRNA, facilitating transfection and intracellular delivery. This review discusses the pathophysiological mechanisms of neurological disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), Huntington's disease (HD), ischemic stroke, spinal cord injury, and Friedreich's ataxia. Additionally, it explores the potential of LNP-mediated mRNA delivery as a therapeutic strategy for these diseases. Various approaches to overcoming BBB-related challenges and enhancing the delivery and efficacy of mRNA-LNPs are discussed, including non-invasive methods with strong potential for clinical translation. With advancements in artificial intelligence (AI)-guided mRNA and LNP design, targeted delivery, gene editing, and CAR-T cell therapy, mRNA-LNPs could significantly transform the treatment landscape for NDD, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Seyedeh Ghazal Moosavi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Zhang H, Liu J, Chen B, Chen X, Wei S, Zhang G, Yan X, Xue X, He G, Lin J, Feng H, Chu W. Human Amniotic Epithelial Stem Cells Promote Functional Recovery After Spinal Cord Injury In Rats By Regulating The Polarization Of Macrophages. Mol Neurobiol 2025; 62:4617-4630. [PMID: 39470871 DOI: 10.1007/s12035-024-04539-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/07/2024] [Indexed: 11/01/2024]
Abstract
Spinal cord injury (SCI) is a catastrophic nerve injury caused by extremely severe damage to the spinal cord, for which effective treatments are currently unavailable. Human amniotic epithelial stem cells (hAESCs) are considered promising candidates for transplantation in various clinical and preclinical applications, due to their lack of limitations such as ethical barriers, immune rejection, tumorigenicity, or cell origin. Nevertheless, the effectiveness and mechanism by which hAESCs treat SCI remain elusive. To assess the motor function recovery process following SCI in rats, the Basso Beattie Bresnahan (BBB) behavior test, inclined plate scale and motor evoked potential (MEP) analysis were used in this study after transplantation of hAESCs at different doses. And the underlying mechanism was investigated by histological and molecular methods. The transplantation of hAESCs can significantly promote the recovery of motor function in SCI group, and the higher the dose, the better the effect. Compared with SCI group, hAESCs group had reduced tissue damage, significantly increased the number of neurons, neurofilaments and myelin sheath, and significantly reduced syringomyelia and glial scars. In addition, hAESCs inhibited the Levels of tumor necrosis factor (TNF-α) and interleukin-6 (IL-6) and increased the expression of the interleukin-4 (IL-4), interleukin-10 (IL-10) and interleukin-13 (IL-13), and promoted the shift of M1-polarized macrophages to M2-polarized macrophages. Our results demonstrate that hAESCs promoted the recovery of motor function after SCI by promoting M2 polarization of macrophages and reducing neuroinflammation. These findings may provide novel therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Hongyan Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jingjing Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Beike Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shijun Wei
- Chongqing Stem Cell and Neural Regeneration Engineering Technology Research Center, Chongqing, 508216, China
| | - Guanghui Zhang
- Chongqing Stem Cell and Neural Regeneration Engineering Technology Research Center, Chongqing, 508216, China
| | - Xiaomin Yan
- Chongqing Stem Cell and Neural Regeneration Engineering Technology Research Center, Chongqing, 508216, China
| | - Xingsen Xue
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Guangjian He
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiangkai Lin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weihua Chu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
3
|
Gong Z, Chen Z, Li D, Lu X, Wu J, Sun H, Wang X, Liu S, Xia X, Lu F, Jiang J, Sun C, Wang H, Zeng F, Ma X. Hydrogel loaded with cerium-manganese nanoparticles and nerve growth factor enhances spinal cord injury repair by modulating immune microenvironment and promoting neuronal regeneration. J Nanobiotechnology 2025; 23:29. [PMID: 39833803 PMCID: PMC11748312 DOI: 10.1186/s12951-025-03098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) treatment remains a formidable challenge, as current therapeutic approaches provide only marginal relief and fail to reverse the underlying tissue damage. This study aims to develop a novel composite material combining enzymatic nanoparticles and nerve growth factor (NGF) to modulate the immune microenvironment and enhance SCI repair. METHODS CeMn nanoparticles (NP) and CeMn NP-polyethylene glycol (PEG) nanozymes were synthesized via sol-gel reaction and DSPE-mPEG modification. Transmission Electron Microscopy, Selected-Area Electron Diffraction, X-ray Diffraction and X-ray Photoelectron Spectroscopy confirmed their crystalline structure, mixed-valence states, and redox properties. Size uniformity, biocompatibility, and catalytic activity were assessed via hydrodynamic diameter, zeta potential, and elemental analysis. The Lightgel/NGF/CeMn NP-PEG composite was synthesized and characterized via electron microscopy, compression testing, rheological analysis, NGF release kinetics, and 30-day degradation studies. Both in vitro and in vivo experiments were conducted to evaluate the therapeutic effects of the composite on SCI. RESULTS The Lightgel/NGF/CeMn NP-PEG composite was successfully synthesized, exhibiting favorable physical properties. At a CeMn NP-PEG concentration of 4 µg/mL, the composite maintained cell viability and demonstrated enhanced biological activity. It also showed superior mechanical properties and an effective NGF release profile. Notably, the composite significantly upregulated the expression of nerve growth-associated proteins, reduced inflammatory cytokines, scavenged reactive oxygen species (ROS), and promoted M2 macrophage polarization by inhibiting the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. In a rat SCI model, it facilitated functional recovery and attenuated inflammation. CONCLUSION The Lightgel/NGF/CeMn NP-PEG composite shows significant therapeutic promise for SCI, effectively eliminating ROS, promoting M2 macrophage polarization, reducing pro-inflammatory cytokines, and supporting neuronal regeneration. These effects substantially enhance motor function in SCI rats, positioning it as a promising candidate for future clinical applications.
Collapse
Affiliation(s)
- Zhaoyang Gong
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Zhenhao Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Dachuan Li
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Xiao Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Jianwei Wu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Hanqiu Sun
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Ximeng Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Siyang Liu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Xinlei Xia
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Feizhou Lu
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Jianyuan Jiang
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Chi Sun
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| | - Hongli Wang
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| | - Feng Zeng
- Artemisinin Research Center, Institute of Science and Technology, The First Affiliated Hospital, The First Clinical Medical School, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510450, China.
| | - Xiaosheng Ma
- Department of Orthopedics, Huashan Hospital, Fudan University, No. 12, Middle Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| |
Collapse
|
4
|
Ma D, Fu C, Li F, Ruan R, Lin Y, Li X, Li M, Zhang J. Functional biomaterials for modulating the dysfunctional pathological microenvironment of spinal cord injury. Bioact Mater 2024; 39:521-543. [PMID: 38883317 PMCID: PMC11179178 DOI: 10.1016/j.bioactmat.2024.04.015] [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: 01/13/2024] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 06/18/2024] Open
Abstract
Spinal cord injury (SCI) often results in irreversible loss of sensory and motor functions, and most SCIs are incurable with current medical practice. One of the hardest challenges in treating SCI is the development of a dysfunctional pathological microenvironment, which mainly comprises excessive inflammation, deposition of inhibitory molecules, neurotrophic factor deprivation, glial scar formation, and imbalance of vascular function. To overcome this challenge, implantation of functional biomaterials at the injury site has been regarded as a potential treatment for modulating the dysfunctional microenvironment to support axon regeneration, remyelination at injury site, and functional recovery after SCI. This review summarizes characteristics of dysfunctional pathological microenvironment and recent advances in biomaterials as well as the technologies used to modulate inflammatory microenvironment, regulate inhibitory microenvironment, and reshape revascularization microenvironment. Moreover, technological limitations, challenges, and future prospects of functional biomaterials to promote efficient repair of SCI are also discussed. This review will aid further understanding and development of functional biomaterials to regulate pathological SCI microenvironment.
Collapse
Affiliation(s)
- Dezun Ma
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Changlong Fu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Fenglu Li
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Renjie Ruan
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| | - Yanming Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Xihai Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Fuzhou, Fujian, 350122, PR China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Min Li
- Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center, 966 Hengyu Road, Fuzhou, 350014, PR China
- Fujian Maternity and Child Health Hospital, 111 Daoshan Road, Fuzhou, 350005, PR China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 111 Daoshan Road, Fuzhou, 350005, PR China
| | - Jin Zhang
- College of Chemical Engineering, Fuzhou University, 2 Xueyuan Road, Fuzhou, 350108, PR China
- Qingyuan Innovation Laboratory, 1 Xueyuan Road, Quanzhou, 362801, PR China
| |
Collapse
|
5
|
Ramos Ferrer P, Vardhan S, Sakiyama-Elbert S. Sustained neurotrophin-3 delivery from hyaluronic acid hydrogels for neural tissue regeneration. J Biomed Mater Res A 2024; 112:1188-1199. [PMID: 37675824 DOI: 10.1002/jbm.a.37596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023]
Abstract
The goal of this work was to design a polymer-based platform capable of localized, long-term delivery of biologically active neurotropic factors using an affinity-based approach. Here, we synthesized hyaluronic acid-methylfuran (HA-mF) hydrogels that provide sustained, affinity-based release of neurotrophin-3 (NT-3), a growth factor that promotes axon growth for 28 days. A Diels-Alder crosslinking reaction between HA-mF and polyethylene glycol (PEG)-dimaleimide occurs within 15 min under physiological conditions, resulting in hydrogels that can be polymerized in the presence of cells and growth factors. We also tuned the hydrogel's storage modulus to match that of native rat spinal cord tissue, providing a platform not only for localized drug delivery but also a suitable vehicle for cellular transplantation. The NT-3 released from the HAmF hydrogels remains bioactive for at least 14 days, promoting axonal growth from primary sensory neurons as well as stem cell-derived V2a interneurons and motoneurons in vitro. The hydrogels also supported cell growth allowing for 3-dimensional axonal extensions within the scaffold matrix. Here we confirm the protective role of HA-mF on matrix-bound NT-3 activity and show that these hydrogels are an excellent platform for growth factor delivery for neural applications.
Collapse
Affiliation(s)
- Pablo Ramos Ferrer
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA
| | - Sangamithra Vardhan
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
6
|
Han X, Zhang M, Yan L, Fu Y, Kou H, Shang C, Wang J, Liu H, Jiang C, Wang J, Cheng T. Role of dendritic cells in spinal cord injury. CNS Neurosci Ther 2024; 30:e14593. [PMID: 38528832 PMCID: PMC10964036 DOI: 10.1111/cns.14593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/15/2023] [Accepted: 12/10/2023] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Inflammation can worsen spinal cord injury (SCI), with dendritic cells (DCs) playing a crucial role in the inflammatory response. They mediate T lymphocyte differentiation, activate microglia, and release cytokines like NT-3. Moreover, DCs can promote neural stem cell survival and guide them toward neuron differentiation, positively impacting SCI outcomes. OBJECTIVE This review aims to summarize the role of DCs in SCI-related inflammation and identify potential therapeutic targets for treating SCI. METHODS Literature in PubMed and Web of Science was reviewed using critical terms related to DCs and SCI. RESULTS The study indicates that DCs can activate microglia and astrocytes, promote T-cell differentiation, increase neurotrophin release at the injury site, and subsequently reduce secondary brain injury and enhance functional recovery in the spinal cord. CONCLUSIONS This review highlights the repair mechanisms of DCs and their potential therapeutic potential for SCI.
Collapse
Affiliation(s)
- Xiaonan Han
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Mingkang Zhang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Liyan Yan
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yikun Fu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongwei Kou
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chunfeng Shang
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Hongjian Liu
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Chao Jiang
- Department of NeurologyThe Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jian Wang
- Department of Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Tian Cheng
- Department of OrthopaedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
7
|
Ertlen C, Seblani M, Bonnet M, Brezun JM, Coyle T, Sabatier F, Fuentes S, Decherchi P, Serratrice N, Marqueste T. Efficacy of the immediate adipose-derived stromal vascular fraction autograft on functional sensorimotor recovery after spinal cord contusion in rats. Stem Cell Res Ther 2024; 15:29. [PMID: 38303017 PMCID: PMC10835949 DOI: 10.1186/s13287-024-03645-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/23/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Spinal cord injuries (SCI) lead to functional alteration with important consequences such as motor and sensory disorders. The repair strategies developed to date remain ineffective. The adipose tissue-derived stromal vascular fraction (SVF) is composed of a cocktail of cells with trophic, pro-angiogenic and immunomodulatory effects. Numerous therapeutic benefits were shown for tissue reconstitution, peripheral neuropathy and for the improvement of neurodegenerative diseases. Here, the therapeutic efficacy of SVF on sensorimotor recovery after an acute thoracic spinal cord contusion in adult rats was determined. METHOD Male Sprague Dawley rats (n = 45) were divided into 3 groups: SHAM (without SCI and treatment), NaCl (animals with a spinal lesion and receiving a saline injection through the dura mater) and SVF (animals with a spinal lesion and receiving a fraction of fat removed from adipocytes through the dura mater). Some animals were sacrificed 14 days after the start of the experiment to determine the inflammatory reaction by measuring the interleukin-1β, interleukin-6 and Tumor Necrosis Factor-α in the lesion area. Other animals were followed once a week for 12 weeks to assess functional recovery (postural and locomotor activities, sensorimotor coordination). At the end of this period, spinal reflexivity (rate-dependent depression of the H-reflex) and physiological adjustments (ventilatory response to metabosensitive muscle activation following muscle fatigue) were measured with electrophysiological tools. RESULTS Compared to non-treated animals, results indicated that the SVF reduced the endogenous inflammation and increased the behavioral recovery in treated animals. Moreover, H-reflex depression and ventilatory adjustments to muscle fatigue were found to be comparable between SHAM and SVF groups. CONCLUSION Our results highlight the effectiveness of SVF and its high therapeutic potential to improve sensorimotor functions and to restore the segmental sensorimotor loop and the communication between supra- and sub-lesional spinal cord regions after traumatic contusion.
Collapse
Affiliation(s)
- Céline Ertlen
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Mostafa Seblani
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Maxime Bonnet
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Jean-Michel Brezun
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Thelma Coyle
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
| | - Florence Sabatier
- Assistance Publique - Hôpitaux de Marseille (AP-HM), INSERM 1409 Centre d'Investigation Clinique en Biothérapies, Unité de Culture Et Thérapie Cellulaire, Hôpital de La Conception, 147, Boulevard Baille, 13385, Marseille Cedex 05, France
| | - Stéphane Fuentes
- Assistance Publique - Hôpitaux de Marseille (AP-HM), Service de Neurochirurgie, Hôpital de La Timone, 264, Rue Saint-Pierre, 13005, Marseille, France
| | - Patrick Decherchi
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France.
| | - Nicolas Serratrice
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France
- Assistance Publique - Hôpitaux de Marseille (AP-HM), Service de Neurochirurgie, Hôpital de La Timone, 264, Rue Saint-Pierre, 13005, Marseille, France
| | - Tanguy Marqueste
- Aix-Marseille Univ, CNRS, ISM UMR 7287, Institut des Sciences du Mouvement: Etienne-Jules MAREY, Equipe Plasticité Des Systèmes Nerveux Et Musculaire (PSNM), Parc Scientifique Et Technologique de Luminy, Aix Marseille Univ, CC910 - 163, Avenue de Luminy, 13288, Marseille Cedex 09, France.
| |
Collapse
|
8
|
Patilas C, Varsamos I, Galanis A, Vavourakis M, Zachariou D, Marougklianis V, Kolovos I, Tsalimas G, Karampinas P, Kaspiris A, Vlamis J, Pneumaticos S. The Role of Interleukin-10 in the Pathogenesis and Treatment of a Spinal Cord Injury. Diagnostics (Basel) 2024; 14:151. [PMID: 38248028 PMCID: PMC10814517 DOI: 10.3390/diagnostics14020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition that often leads to severe and permanent neurological deficits. The complex pathophysiology of an SCI involves a cascade of events, including inflammation, oxidative stress, and secondary injury processes. Among the myriad of molecular players involved, interleukin-10 (IL-10) emerges as a key regulator with the potential to modulate both the inflammatory response and promote neuroprotection. This comprehensive review delves into the intricate interplay of IL-10 in the pathogenesis of an SCI and explores its therapeutic implications in the quest for effective treatments. IL-10 has been found to regulate inflammation, oxidative stress, neuronal apoptosis, and glial scars after an SCI. Its neuroprotective properties have been evaluated in a plethora of animal studies. IL-10 administration, either isolated or in combination with other molecules or biomaterials, has shown neuroprotective effects through a reduction in inflammation, the promotion of tissue repair and regeneration, the modulation of glial scar formation, and improved functional outcomes. In conclusion, IL-10 emerges as a pivotal player in the pathogenesis and treatment of SCIs. Its multifaceted role in modulating inflammation, oxidative stress, neuronal apoptosis, glial scars, and neuroprotection positions IL-10 as a promising therapeutic target. The ongoing research exploring various strategies for harnessing the potential of IL-10 offers hope for the development of effective treatments that could significantly improve outcomes for individuals suffering from spinal cord injuries. As our understanding of IL-10's intricacies deepens, it opens new avenues for innovative and targeted therapeutic interventions, bringing us closer to the goal of alleviating the profound impact of SCIs on patients' lives.
Collapse
Affiliation(s)
| | | | | | - Michail Vavourakis
- 3rd Department of Orthopaedic Surgery, National & Kapodistrian University of Athens, KAT General Hospital, 14561 Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Javanbakht P, Yazdi FR, Taghizadeh F, Khadivi F, Hamidabadi HG, Kashani IR, Zarini D, Mojaverrostami S. Quercetin as a possible complementary therapy in multiple sclerosis: Anti-oxidative, anti-inflammatory and remyelination potential properties. Heliyon 2023; 9:e21741. [PMID: 37954351 PMCID: PMC10638059 DOI: 10.1016/j.heliyon.2023.e21741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder of the central nervous system (CNS) which causes various symptoms such as fatigue, dyscoordination weakness and visual weakness. The intricacy of the immune system and obscure etiology are the main reasons for the lack of a definite treatment for MS. Oxidative stress is one of the most important key factors in MS pathogenesis. It can enhance inflammation, neurodegeneration and autoimmune-mediated processes, which can lead to excessive demyelination and axonal disruption. Recently, promising effects of Quercetin as a non-pharmacological anti-oxidant therapy have been reported in preclinical studies of MS disease. In this review, we provide a compendium of preclinical and clinical studies that have investigated the effects of Quercetin on MS disease to evaluate its potential utility as a complementary therapy in MS. Quercetin treatment in MS disease not only protects the CNS against oxidative stress and neuroinflammation, but it also declines the demyelination process and promotes remyelination potential. The present study clarifies the reported knowledge on the beneficial effects of Quercetin against MS, with future implication as a neuroprotective complementary therapy.
Collapse
Affiliation(s)
- Parinaz Javanbakht
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzane Rezaei Yazdi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Taghizadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Zarini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Li J, Wang P, Zhou T, Jiang W, Wu H, Zhang S, Deng L, Wang H. Neuroprotective effects of interleukin 10 in spinal cord injury. Front Mol Neurosci 2023; 16:1214294. [PMID: 37492521 PMCID: PMC10363608 DOI: 10.3389/fnmol.2023.1214294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Spinal cord injury (SCI) starts with a mechanical and/or bio-chemical insult, followed by a secondary phase, leading progressively to severe collapse of the nerve tissue. Compared to the peripheral nervous system, injured spinal cord is characterized by weak axonal regeneration, which leaves most patients impaired or paralyzed throughout lifetime. Therefore, confining, alleviating, or reducing the expansion of secondary injuries and promoting functional connections between rostral and caudal regions of lesion are the main goals of SCI therapy. Interleukin 10 (IL-10), as a pivotal anti-inflammatory and immunomodulatory cytokine, exerts a wide spectrum of positive effects in the treatment of SCI. The mechanisms underlying therapeutic effects mainly include anti-oxidative stress, limiting excessive inflammation, anti-apoptosis, antinociceptive effects, etc. Furthermore, IL-10 displays synergistic effects when combined with cell transplantation or neurotrophic factor, enhancing treatment outcomes. This review lists pleiotropic mechanisms underlying IL-10-mediated neuroprotection after SCI, which may offer fresh perspectives for clinical translation.
Collapse
Affiliation(s)
- Juan Li
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Pei Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Ting Zhou
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Wenwen Jiang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Hang Wu
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Shengqi Zhang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Lingxiao Deng
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| |
Collapse
|
11
|
Janzadeh A, Ramezani F, Yousefi S, Hamblin MR, Mojarad N, Nasirinezhad F. Time-dependent photobiomodulation management of neuropathic pain induced by spinal cord injury in male rats. Lasers Med Sci 2023; 38:120. [PMID: 37160475 DOI: 10.1007/s10103-023-03722-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/16/2023] [Indexed: 05/11/2023]
Abstract
Neuropathic pain (NP) following spinal cord injury (SCI) often lasts for a long time and causes a range of problems that reduce the quality of life. Current treatments are not generally effective; however, photobiomodulation therapy (PBMT) has made some progress in this area. Due to the novelty of this treatment, standard therapeutic protocols have not yet been agreed upon. In the present study, we compare the analgesic effect of two PBMT protocols (2 and 4 weeks of radiation). A total of thirty-two adult male Wistar rats were divided into four groups: control, SCI, 2 W PBMT, and 4 W PBMT. SCI was induced by an aneurism clip and PBMT used a 660-nm, initiated 30 min post-SCI, and continued daily for 2 or 4 weeks. Functional recovery, hyperalgesia, and allodynia were measured weekly. At the end of the study, the Gad65, interleukin 1-alpha (IL1α), interleukin 10 (IL10), IL4, and purinergic receptor (P2xR and P2yR) expressions were measured. Data were analyzed by Prism6. The results showed PBM irradiation for 2 and 4 weeks had the same effects in improving hyperalgesia. In the case of allodynia and functional recovery, 4 W PBMT was more effective (p<0.01). 4 W PBMT increased the Gad65 expression (p <0.001) and reduced P2Y4R (p <0.05) compared to SCI animals. The effects of 2 and 4 W PBMT were the same for IL1α, IL10, and P2X3 receptors. 4 W PBMT was more effective in reducing the complications of SCI such as pain and disability. PBMT therapy is an effective method aimed at immune system function modulation to reduce NP and motor dysfunction.
Collapse
Affiliation(s)
- Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sogol Yousefi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Michael R Hamblin
- Laser Research Center, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Negin Mojarad
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - Farinaz Nasirinezhad
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Gál L, Bellák T, Marton A, Fekécs Z, Weissman D, Török D, Biju R, Vizler C, Kristóf R, Beattie MB, Lin PJ, Pardi N, Nógrádi A, Pajer K. Restoration of Motor Function through Delayed Intraspinal Delivery of Human IL-10-Encoding Nucleoside-Modified mRNA after Spinal Cord Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0056. [PMID: 36930811 PMCID: PMC10013810 DOI: 10.34133/research.0056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
Efficient in vivo delivery of anti-inflammatory proteins to modulate the microenvironment of an injured spinal cord and promote neuroprotection and functional recovery is a great challenge. Nucleoside-modified messenger RNA (mRNA) has become a promising new modality that can be utilized for the safe and efficient delivery of therapeutic proteins. Here, we used lipid nanoparticle (LNP)-encapsulated human interleukin-10 (hIL-10)-encoding nucleoside-modified mRNA to induce neuroprotection and functional recovery following rat spinal cord contusion injury. Intralesional administration of hIL-10 mRNA-LNP to rats led to a remarkable reduction of the microglia/macrophage reaction in the injured spinal segment and induced significant functional recovery compared to controls. Furthermore, hIL-10 mRNA treatment induced increased expression in tissue inhibitor of matrix metalloproteinase 1 and ciliary neurotrophic factor levels in the affected spinal segment indicating a time-delayed secondary effect of IL-10 5 d after injection. Our results suggest that treatment with nucleoside-modified mRNAs encoding neuroprotective factors is an effective strategy for spinal cord injury repair.
Collapse
Affiliation(s)
- László Gál
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tamás Bellák
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Annamária Marton
- National Biotechnology Laboratory, Institute of Genetics, Biological Research Centre, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Zoltán Fekécs
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dénes Török
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Rachana Biju
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Csaba Vizler
- National Biotechnology Laboratory, Institute of Genetics, Biological Research Centre, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Rebeka Kristóf
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | | | | | - Norbert Pardi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| |
Collapse
|
13
|
Park J, Farmer M, Casson C, Kalashnikova I, Kolpek D. Therapeutic Potential of Combinative shRNA-Encoded Lentivirus-Mediated Gene Silencing to Accelerate Somatosensory Recovery After Spinal Cord Trauma. Neurotherapeutics 2023; 20:564-577. [PMID: 36401079 PMCID: PMC10121969 DOI: 10.1007/s13311-022-01331-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
Neuropathic pain following spinal cord injury (SCI) remains a difficult problem that affects more than 80% of SCI patients. Growing evidence indicates that neuroinflammatory responses play a key role in neuropathic pain after SCI. Short hairpin RNA (shRNA) interference is an efficient tool for the knockdown of disease-related specific gene expression after SCI, yet insufficient data is available to establish guidelines. In this study, we have constructed the transient receptor potential ankyrin 1 (TRPA1) shRNA encoded-lentiviral vector (LV-shTRPA1) and P38 MAPK shRNA encoded-lentiviral vector (LV-shP38) to investigate the silencing effects of shRNAs and their ability to reprogram the neuroinflammatory responses, thereby enhancing somatosensory recovery after SCI. Our in vitro data employing HEK293-FT and activated macrophages demonstrated that delivered LV-shRNAs showed high transduction efficacy with no cytotoxicity. Furthermore, a combination of LV-shP38 and LV-shTRPA1 was found to be most effective at suppressing target genes, cutting the expression of pro-inflammatory and pro-nociceptive factors in the dorsal horn of the spinal cord and dorsal root ganglia, thus contributing to the alleviation of neuronal hypersensitivities after SCI. Overall, our data demonstrated that the combination LV-shP38/shTRPA1 produced a synergistic effect for immunomodulation and reduced neuropathic pain with a favorable risk-to-benefit ratio. Collectively, our LV-mediated shRNA delivery will provide an efficient tool for gene silencing therapeutic approaches to treat various incurable disorders.
Collapse
Affiliation(s)
- Jonghyuck Park
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA.
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone, Lexington, KY, 40506, USA.
| | - Matthew Farmer
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Camara Casson
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Irina Kalashnikova
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Daniel Kolpek
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| |
Collapse
|
14
|
Suzuki H, Imajo Y, Funaba M, Ikeda H, Nishida N, Sakai T. Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24032528. [PMID: 36768846 PMCID: PMC9917245 DOI: 10.3390/ijms24032528] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
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 preclinical research and clinical trials. In the near future, several more are expected to come down the translational pipeline. Among ongoing and completed trials are those reporting the use of biomaterial scaffolds. The advancements in biomaterial technology, combined with stem cell therapy or other regenerative therapy, can now accelerate the progress of promising novel therapeutic strategies from bench to bedside. Various types of approaches to regeneration therapy for SCI have been combined with the use of supportive biomaterial scaffolds as a drug and cell delivery system to facilitate favorable cell-material interactions and the supportive effect of neuroprotection. In this review, we summarize some of the most recent insights of preclinical and clinical studies using biomaterial scaffolds in regenerative therapy for SCI and summarized the biomaterial strategies for treatment with simplified results data. One hundred and sixty-eight articles were selected in the present review, in which we focused on biomaterial scaffolds. We conducted our search of articles using PubMed and Medline, a medical database. We used a combination of "Spinal cord injury" and ["Biomaterial", or "Scaffold"] as search terms and searched articles published up until 30 April 2022. Successful future therapies will require these biomaterial scaffolds and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, the loss of a structural framework, and biocompatibility. This database could serve as a benchmark to progress in future clinical trials for SCI using biomaterial scaffolds.
Collapse
|
15
|
Xia Y, Yang R, Wang H, Hou Y, Li Y, Zhu J, Xu F, Fu C. Biomaterials delivery strategies to repair spinal cord injury by modulating macrophage phenotypes. J Tissue Eng 2022; 13:20417314221143059. [PMID: 36600997 PMCID: PMC9806413 DOI: 10.1177/20417314221143059] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/17/2022] [Indexed: 12/28/2022] Open
Abstract
Spinal cord injury (SCI) causes tremendous harm to a patient's physical, mental, and financial health. Moreover, recovery of SCI is affected by many factors, inflammation is one of the most important as it engulfs necrotic tissue and cells during the early stages of injury. However, excessive inflammation is not conducive to damage repair. Macrophages are classified into either blood-derived macrophages or resident microglia based on their origin, their effects on SCI being two-sided. Microglia first activate and recruit blood-derived macrophages at the site of injury-blood-borne macrophages being divided into pro-inflammatory M1 phenotypes and anti-inflammatory M2 phenotypes. Among them, M1 macrophages secrete inflammatory factors such as interleukin-β (IL-β), tumor necrosis factor-α (TNF-α), IL-6, and interferon-γ (IFN-γ) at the injury site, which aggravates SCIs. M2 macrophages secrete IL-4, IL-10, IL-13, and neurotrophic factors to inhibit the inflammatory response and inhibit neuronal apoptosis. Consequently, modulating phenotypic differentiation of macrophages appears to be a meaningful therapeutic target for the treatment of SCI. Biomaterials are widely used in regenerative medicine and tissue engineering due to their targeting and bio-histocompatibility. In this review, we describe the effects of biomaterials applied to modulate macrophage phenotypes on SCI recovery and provide an outlook.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Ruohan Yang
- Cancer Center, The First Hospital of
Jilin University, Changchun, PR China
| | - Hengyi Wang
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Yulin Hou
- Depattment of Cardiology, Guangyuan
Central Hospital, Guangyuan, PR China
| | - Yuehong Li
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Feng Xu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China
| | - Changfeng Fu
- Department of Spine Surgery, The First
Hospital of Jilin University, Changchun, PR China,Changfeng Fu, Department of Spine Surgery,
The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR
China.
| |
Collapse
|
16
|
Ciciriello AJ, Surnar B, Medy GD, Su X, Dhar S, Dumont CM. Biomaterial-targeted precision nanoparticle delivery to the injured spinal cord. Acta Biomater 2022; 152:532-545. [PMID: 36087868 PMCID: PMC10551882 DOI: 10.1016/j.actbio.2022.08.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 11/01/2022]
Abstract
Drug delivery requires precision in timing, location, and dosage to achieve therapeutic benefits. Challenges in addressing all three of these critical criteria result in poor temporal dexterity, widespread accumulation and off-target effects, and high doses with the potential for toxicity. To address these challenges, we have developed the BiomatErial Accumulating Carriers for On-demand Nanotherapy (BEACON) platform that utilizes an implantable biomaterial to serve as a target for systemically delivered nanoparticles (NPs). With the BEACON system, administered NPs are conjugated with a ligand that has high affinity for a receptor in the implanted biomaterial. To test BEACON, an in vivo spinal cord injury (SCI) model was used as it provides an injury model where the three identified criteria can be tested as it is a dynamic and complicated injury model with no currently approved therapies. Through our work, we have demonstrated temporal dexterity in NP administration by injecting 6 days post-SCI, decreased off-target accumulation with a significant drop in liver accumulation, and retention of our NPs in the target biomaterial. The BEACON system can be applied broadly, beyond the nervous system, to improve systemically delivered NP accumulation at an implanted biomaterial target. STATEMENT OF SIGNIFICANCE: Targeted drug delivery approaches have the potential to improve therapeutic regimens for patients on a case-by-case basis. Improved localization of a therapeutic to site of interest can result in increased efficacy and limit the need for repeat dosing. Unfortunately, targeted strategies can fall short when receptors on cells or tissues are too widespread or change over the course of disease or injury progression. The BEACON system developed herein eliminates the need to target a cell or tissue receptor by targeting an implantable biomaterial with location-controllable accumulation and sustained presentation over time. The targeting paradigm presented by BEACON is widely applicable throughout tissue engineering and regenerative medicine without the need to retool for each new application.
Collapse
Affiliation(s)
- Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States
| | - Bapurao Surnar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Giovanni D Medy
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Xiaoyu Su
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Shanta Dhar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami, 1475 NW 12th Avenue, Miami, Florida 33136, United States
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States.
| |
Collapse
|
17
|
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
|
18
|
Gao T, Huang F, Wang W, Xie Y, Wang B. Interleukin-10 genetically modified clinical-grade mesenchymal stromal cells markedly reinforced functional recovery after spinal cord injury via directing alternative activation of macrophages. Cell Mol Biol Lett 2022; 27:27. [PMID: 35300585 PMCID: PMC8931978 DOI: 10.1186/s11658-022-00325-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Background After spinal cord injury (SCI), dysregulated or nonresolving inflammatory processes can severely disturb neuronal homeostasis and drive neurodegeneration. Although mesenchymal stromal cell (MSC)-based therapies have showed certain therapeutic efficacy, no MSC therapy has reached its full clinical goal. In this study, we examine interleukin-10 (IL10) genetically modified clinical-grade MSCs (IL10-MSCs) and evaluate their clinical safety, effectiveness, and therapeutic mechanism in a completely transected SCI mouse model. Methods We established stable IL10-overexpressing human umbilical-cord-derived MSCs through electric transduction and screened out clinical-grade IL10-MSCs according to the criteria of cell-based therapeutic products, which were applied to mice with completely transected SCI by repeated tail intravenous injections. Then we comprehensively investigated the motor function, histological structure, and nerve regeneration in SCI mice, and further explored the potential therapeutic mechanism after IL10-MSC treatment. Results IL10-MSC treatment markedly reinforced locomotor improvement, accompanied with decreased lesion volume, regeneration of axons, and preservation of neurons, compared with naïve unmodified MSCs. Further, IL10-MSC transplantation increased the ratio of microglia to infiltrated alternatively activated macrophages (M2), and reduced the ratio of classically activated macrophages (M1) at the injured spinal cord, meanwhile increasing the percentage of Treg and Th2 cells, and reducing the percentage of Th1 cells in the peripheral circulatory system. In addition, IL10-MSC administration could prevent apoptosis and promote neuron differentiation of neural stem cells (NSCs) under inflammatory conditions in vitro. Conclusions IL10-MSCs exhibited a reliable safety profile and demonstrated promising therapeutic efficacy in SCI compared with naïve MSCs, providing solid support for future clinical application of genetically engineered MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00325-9.
Collapse
Affiliation(s)
- Tianyun Gao
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Feifei Huang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Wenqing Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuanyuan Xie
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
19
|
Tan Z, Yang G, Qiu J, Yan W, Liu Y, Ma Z, Li J, Liu J, Shan N. Quercetin Alleviates Demyelination Through Regulating Microglial Phenotype Transformation to Mitigate Neuropsychiatric Symptoms in Mice with Vascular Dementia. Mol Neurobiol 2022; 59:3140-3158. [PMID: 35267135 DOI: 10.1007/s12035-021-02712-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022]
Abstract
Cerebral hypoperfusion plays a pivotal role in the ictus and development of vascular dementia (VaD) with neuropsychiatric symptoms. To date, few pharmacological interventions for neuropsychiatric symptoms are available in the VaD patients with neuropsychiatric impairments. Here, our results demonstrated that the extent of demyelination was dramatically deteriorated and the thickness of myelin sheath was evidently decreased in the presence of cerebral hypoperfusion, whereas Quercetin possessed the potential of abrogating these effects at least in part, then relieving anxiety and depression-like behavior when mice exposed to bilateral carotid artery stenosis (BCAS)/chronic restraint stress (CRS). The underlying mechanism was that Quercetin facilitated secretion of anti-inflammatory cytokines (IL-4 and IL-10) and in turn decreased production of pro-inflammatory factors (TNF-α and IL-1β) due to regulating microglial phenotype transformation, thereafter enhancing the microglial engulfment ability of myelin fragments in vitro and in vivo. Collectively, the results demonstrated that that Quercetin mediated microglial transformation into anti-inflammatory phenotype to reduce demyelination in ventral hippocampus (vHIP), thereafter mitigating neuropsychiatric deficits (including anxiety and depression). The present research broadens the therapeutic scope of Quercetin in central nervous system (CNS) disorders with presence of white matter damage and/or the insufficient activation of anti-inflammatory microglia, particularly for vascular dementia with/without neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Zihu Tan
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Guang Yang
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Jing Qiu
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Wenjing Yan
- Clinical College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Yu Liu
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Zhengling Ma
- Clinical College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Jia Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment By Acupuncture and Moxibustion, Wuhan, 430061, China
| | - Jing Liu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment By Acupuncture and Moxibustion, Wuhan, 430061, China
| | - Nan Shan
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China. .,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China. .,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
20
|
Dervan A, Franchi A, Almeida-Gonzalez FR, Dowling JK, Kwakyi OB, McCoy CE, O’Brien FJ, Hibbitts A. Biomaterial and Therapeutic Approaches for the Manipulation of Macrophage Phenotype in Peripheral and Central Nerve Repair. Pharmaceutics 2021; 13:2161. [PMID: 34959446 PMCID: PMC8706646 DOI: 10.3390/pharmaceutics13122161] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/18/2022] Open
Abstract
Injury to the peripheral or central nervous systems often results in extensive loss of motor and sensory function that can greatly diminish quality of life. In both cases, macrophage infiltration into the injury site plays an integral role in the host tissue inflammatory response. In particular, the temporally related transition of macrophage phenotype between the M1/M2 inflammatory/repair states is critical for successful tissue repair. In recent years, biomaterial implants have emerged as a novel approach to bridge lesion sites and provide a growth-inductive environment for regenerating axons. This has more recently seen these two areas of research increasingly intersecting in the creation of 'immune-modulatory' biomaterials. These synthetic or naturally derived materials are fabricated to drive macrophages towards a pro-repair phenotype. This review considers the macrophage-mediated inflammatory events that occur following nervous tissue injury and outlines the latest developments in biomaterial-based strategies to influence macrophage phenotype and enhance repair.
Collapse
Affiliation(s)
- Adrian Dervan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Antonio Franchi
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Francisco R. Almeida-Gonzalez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Jennifer K. Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Ohemaa B. Kwakyi
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- School of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Claire E. McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Alan Hibbitts
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| |
Collapse
|
21
|
A DAMP-scavenging, IL-10-releasing hydrogel promotes neural regeneration and motor function recovery after spinal cord injury. Biomaterials 2021; 280:121279. [PMID: 34847433 DOI: 10.1016/j.biomaterials.2021.121279] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022]
Abstract
Spinal cord injury (SCI) creates an inflammatory microenvironment characterized by damage-associated molecular patterns (DAMPs) and immune cell activation that exacerbate secondary damage and impair neurological recovery. Here we develop an immunoregulatory hydrogel scaffold for treating SCI that scavenges DAMPs and slowly releases the anti-inflammatory cytokine interleukin-10 (IL-10). We created this dual-functional scaffold by modifying a photocrosslinked gelatin hydrogel with the cationic, DAMP-binding polymer poly (amidoamine) and with IL-10, and compared the therapeutic activity of this scaffold with that of gelatin-only, gelatin + poly (amidoamine), and gelatin + IL-10 scaffolds in vitro and in vivo. In vitro, the dual-functional scaffold scavenged anionic DAMPs and exhibited sustained release of IL-10, reduced the proinflammatory responses of macrophages and microglia, and enhanced the neurogenic differentiation of neural stem cells. In a complete transection SCI mouse model, the injected dual-functional scaffold suppressed proinflammatory cytokine production, promoted the M2 macrophage/microglia phenotype, and led to neural regeneration and axon growth without scar formation to a greater extent than the single-function or control scaffolds. This DAMP-scavenging, IL-10-releasing scaffold provides a new strategy for promoting neural regeneration and motor function recovery following severe SCI.
Collapse
|
22
|
Li Z, Wang Q, Hu H, Zheng W, Gao C. Research advances of biomaterials-based microenvironment-regulation therapies for repair and regeneration of spinal cord injury. Biomed Mater 2021; 16. [PMID: 34384071 DOI: 10.1088/1748-605x/ac1d3c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022]
Abstract
Traumatic spinal cord injury (SCI) usually results in restricted behaviour recovery and even life-changing paralysis, accompanied with numerous complications. Pathologically, the initial injuries trigger a series of secondary injuries, leading to an expansion of lesion site, a mass of neuron loss, and eventual failure of endogenous axon regeneration. As the advances rapidly spring up in regenerative medicine and tissue engineering biomaterials, regulation of these secondary injuries becomes possible, shedding a light on normal functional restoration. The successful tissue regeneration lies in proper regulation of the inflammatory microenvironment, including the inflammatory immune cells and inflammatory factors that lead to oxidative stress, inhibitory glial scar and neuroexcitatory toxicity. Specifically, the approaches based on microenvironment-regulating biomaterials have shown great promise in the repair and regeneration of SCI. In this review, the pathological inflammatory microenvironments of SCI are discussed, followed by the introduction of microenvironment-regulating biomaterials in terms of their impressive therapeutic effect in attenuation of secondary inflammation and promotion of axon regrowth. With the emphasis on regulating secondary events, the biomaterials for SCI treatment will become promising for clinical applications.
Collapse
Affiliation(s)
- Ziming Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China.,Dr Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
23
|
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
|
24
|
Sun WM, Ma CL, Xu J, He JP. Reduction in post-spinal cord injury spasticity by combination of peripheral nerve grafting and acidic fibroblast growth factor infusion in monkeys. J Int Med Res 2021; 49:3000605211022294. [PMID: 34154433 PMCID: PMC8236803 DOI: 10.1177/03000605211022294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Spasticity is a frequent complication after spinal cord injury (SCI), but the existing therapies provide only limited relief and are associated with adverse reactions. Therefore, we aimed to develop a novel strategy to ameliorate the spasticity induced by SCI. METHODS This nonrandomized controlled study used a repeated measurement design. The study involved four monkeys, two of which served as controls and only underwent spinal cord hemisection surgery at the T8 spine level. The other two monkeys underwent transplantation of sural nerve segments into the injured sites and long-term infusion of acidic fibroblast growth factor (aFGF). All monkeys received postoperative exercise training and therapy. RESULTS The combined therapy substantially reduced the spasticity in leg muscle tone, patella tendon reflex, and fanning of toes. Although all monkeys showed spontaneous recovery of function over time, the recovery in the controls reached a plateau and started to decline after 11 weeks. CONCLUSIONS The combination of peripheral nerve grafting and aFGF infusion may serve as a complementary approach to reduce the signs of spasticity in patients with SCI.
Collapse
Affiliation(s)
- Wei-Ming Sun
- Institute of Life Science, Nanchang University, Nanchang, China.,School of Life Science, Nanchang University, Nanchang, China
| | - Chao-Lin Ma
- Institute of Life Science, Nanchang University, Nanchang, China.,School of Life Science, Nanchang University, Nanchang, China
| | - Jiang Xu
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Ping He
- Center for Neural Interface Design, School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
25
|
Tejeda G, Ciciriello AJ, Dumont CM. Biomaterial Strategies to Bolster Neural Stem Cell-Mediated Repair of the Central Nervous System. Cells Tissues Organs 2021; 211:655-669. [PMID: 34120118 DOI: 10.1159/000515351] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/12/2021] [Indexed: 01/25/2023] Open
Abstract
Stem cell therapies have the potential to not only repair, but to regenerate tissue of the central nervous system (CNS). Recent studies demonstrate that transplanted stem cells can differentiate into neurons and integrate with the intact circuitry after traumatic injury. Unfortunately, the positive findings described in rodent models have not been replicated in clinical trials, where the burden to maintain the cell viability necessary for tissue repair becomes more challenging. Low transplant survival remains the greatest barrier to stem cell-mediated repair of the CNS, often with fewer than 1-2% of the transplanted cells remaining after 1 week. Strategic transplantation parameters, such as injection location, cell concentration, and transplant timing achieve only modest improvements in stem cell transplant survival and appear inconsistent across studies. Biomaterials provide researchers with a means to significantly improve stem cell transplant survival through two mechanisms: (1) a vehicle to deliver and protect the stem cells and (2) a substrate to control the cytotoxic injury environment. These biomaterial strategies can alleviate cell death associated with delivery to the injury and can be used to limit cell death after transplantation by limiting cell exposure to cytotoxic signals. Moreover, it is likely that control of the injury environment with biomaterials will lead to a more reliable support for transplanted cell populations. This review will highlight the challenges associated with cell delivery in the CNS and the advances in biomaterial development and deployment for stem cell therapies necessary to bolster stem cell-mediated repair.
Collapse
Affiliation(s)
- Giancarlo Tejeda
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| |
Collapse
|
26
|
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
|
27
|
Abstract
Traumatic spinal cord injury (SCI) results in direct and indirect damage to neural tissues, which results in motor and sensory dysfunction, dystonia, and pathological reflex that ultimately lead to paraplegia or tetraplegia. A loss of cells, axon regeneration failure, and time-sensitive pathophysiology make tissue repair difficult. Despite various medical developments, there are currently no effective regenerative treatments. Stem cell therapy is a promising treatment for SCI due to its multiple targets and reactivity benefits. The present review focuses on SCI stem cell therapy, including bone marrow mesenchymal stem cells, umbilical mesenchymal stem cells, adipose-derived mesenchymal stem cells, neural stem cells, neural progenitor cells, embryonic stem cells, induced pluripotent stem cells, and extracellular vesicles. Each cell type targets certain features of SCI pathology and shows therapeutic effects via cell replacement, nutritional support, scaffolds, and immunomodulation mechanisms. However, many preclinical studies and a growing number of clinical trials found that single-cell treatments had only limited benefits for SCI. SCI damage is multifaceted, and there is a growing consensus that a combined treatment is needed.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, 34753West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Xiong
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| |
Collapse
|
28
|
Bai YR, Lai BQ, Han WT, Sun JH, Li G, Ding Y, Zeng X, Ma YH, Zeng YS. Decellularized optic nerve functional scaffold transplant facilitates directional axon regeneration and remyelination in the injured white matter of the rat spinal cord. Neural Regen Res 2021; 16:2276-2283. [PMID: 33818513 PMCID: PMC8354131 DOI: 10.4103/1673-5374.310696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Axon regeneration and remyelination of the damaged region is the most common repair strategy for spinal cord injury. However, achieving good outcome remains difficult. Our previous study showed that porcine decellularized optic nerve better mimics the extracellular matrix of the embryonic porcine optic nerve and promotes the directional growth of dorsal root ganglion neurites. However, it has not been reported whether this material promotes axonal regeneration in vivo. In the present study, a porcine decellularized optic nerve was seeded with neurotrophin-3-overexpressing Schwann cells. This functional scaffold promoted the directional growth and remyelination of regenerating axons. In vitro, the porcine decellularized optic nerve contained many straight, longitudinal channels with a uniform distribution, and microscopic pores were present in the channel wall. The spatial micro topological structure and extracellular matrix were conducive to the adhesion, survival and migration of neural stem cells. The scaffold promoted the directional growth of dorsal root ganglion neurites, and showed strong potential for myelin regeneration. Furthermore, we transplanted the porcine decellularized optic nerve containing neurotrophin-3-overexpressing Schwann cells in a rat model of T10 spinal cord defect in vivo. Four weeks later, the regenerating axons grew straight, the myelin sheath in the injured/transplanted area recovered its structure, and simultaneously, the number of inflammatory cells and the expression of chondroitin sulfate proteoglycans were reduced. Together, these findings suggest that porcine decellularized optic nerve loaded with Schwann cells overexpressing neurotrophin-3 promotes the directional growth of regenerating spinal cord axons as well as myelin regeneration. All procedures involving animals were conducted in accordance with the ethical standards of the Institutional Animal Care and Use Committee of Sun Yat-sen University (approval No. SYSU-IACUC-2019-B034) on February 28, 2019.
Collapse
Affiliation(s)
- Yu-Rong Bai
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wei-Tao Han
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jia-Hui Sun
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Institute of Spinal Cord Injury; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
29
|
Chen JY, Fu EJ, Patel PR, Hostetler AJ, Sawan HA, Moss KA, Hocevar SE, Anderson AJ, Chestek CA, Shea LD. Lentiviral Interleukin-10 Gene Therapy Preserves Fine Motor Circuitry and Function After a Cervical Spinal Cord Injury in Male and Female Mice. Neurotherapeutics 2021; 18:503-514. [PMID: 33051853 PMCID: PMC8116384 DOI: 10.1007/s13311-020-00946-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2020] [Indexed: 12/16/2022] Open
Abstract
In mammals, spinal cord injuries often result in muscle paralysis through the apoptosis of lower motor neurons and denervation of neuromuscular junctions. Previous research shows that the inflammatory response to a spinal cord injury can cause additional tissue damage after the initial trauma. To modulate this inflammatory response, we delivered lentiviral anti-inflammatory interleukin-10, via loading onto an implantable biomaterial scaffold, into a left-sided hemisection at the C5 vertebra in mice. We hypothesized that improved behavioral outcomes associated with anti-inflammatory treatment are due to the sparing of fine motor circuit components. We examined behavioral recovery using a ladder beam, tissue sparing using histology, and electromyogram recordings using intraspinal optogenetic stimulation at 2 weeks post-injury. Ladder beam analysis shows interleukin-10 treatment results in significant improvement of behavioral recovery at 2 and 12 weeks post-injury when compared to mice treated with a control virus. Histology shows interleukin-10 results in greater numbers of lower motor neurons, axons, and muscle innervation at 2 weeks post-injury. Furthermore, electromyogram recordings suggest that interleukin-10-treated animals have signal-to-noise ratios and peak-to-peak amplitudes more similar to that of uninjured controls than to that of control injured animals at 2 weeks post-injury. These data show that gene therapy using anti-inflammatory interleukin-10 can significantly reduce tissue damage and subsequent motor deficits after a spinal cord injury. Together, these results suggest that early modulation of the injury response can preserve muscle function with long-lasting benefits.
Collapse
Affiliation(s)
- Jessica Y Chen
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Emily J Fu
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
| | - Paras R Patel
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
| | - Alexander J Hostetler
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
| | - Hasan A Sawan
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
| | - Kayla A Moss
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
| | - Sarah E Hocevar
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
- Robotics Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Boulevard, 1119 Carl A Gerstacker Building, Ann Arbor, MI, 48109, USA.
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
30
|
Ciciriello AJ, Smith DR, Munsell MK, Boyd SJ, Shea LD, Dumont CM. Acute Implantation of Aligned Hydrogel Tubes Supports Delayed Spinal Progenitor Implantation. ACS Biomater Sci Eng 2020; 6:5771-5784. [DOI: 10.1021/acsbiomaterials.0c00844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew J. Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida 33136, United States
| | - Dominique R. Smith
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
| | - Mary K. Munsell
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
| | - Sydney J. Boyd
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, Michigan 48109, United States
| | - Courtney M. Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida 33136, United States
| |
Collapse
|