Next Article in Journal
Pathway and Network Analyses Identify Growth Factor Signaling and MMP9 as Potential Mediators of Mitochondrial Dysfunction in Severe COVID-19
Previous Article in Journal
Consistency and Variation in Doublecortin and Ki67 Antigen Detection in the Brain Tissue of Different Mammals, including Humans
Previous Article in Special Issue
Bacterial Cellulose-Based Blends and Composites: Versatile Biomaterials for Tissue Engineering Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury

Department of Orthopedics Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2528; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24032528
Submission received: 16 December 2022 / Revised: 5 January 2023 / Accepted: 11 January 2023 / Published: 28 January 2023
(This article belongs to the Special Issue Biomaterials and Regenerative Medicine)

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.

1. Introduction

Spinal cord injuries (SCIs) are a serious problem for those affected. The physical, emotional and economic problems caused by SCI generally considerably limit an individual’s functionality and are a burden on society. One recent survey reported an annual incidence of SCI of approximately 54 cases per one million people [1,2], with an estimated yearly incidence worldwide of 250,000–500,000 cases [3]. The spinal cord has very little ability to spontaneously or functionally regenerate itself, thus resulting in serious and often permanent disabilities. Unfortunately, 95% of patients with SCI are in the chronic phase [4]. The cause of SCI is triggered by several types of physical impacts, including traffic accidents, falls and sports injuries, etc., in which spinal vertebrae, facet joints, disks and ligamentous structures are injured and lose stability. The impact load is therefore transferred to the spinal cord and injures it. The external insult is reflected in primary spinal tissue damage and neural cell death in the acute phase, while a subsequential secondary cascade of degenerative events is started [5] (Figure 1).
Spinal surgery is often necessary to reduce the cervical dislocation and to remove fragments of bones, herniated disks, foreign objects or fractured vertebrae that appear to be compressing the spinal cord and cervical nerve roots. Surgery might also be needed to stabilize the spine to prevent future pain or deformity. Unfortunately, there is no way to reverse damage to the original spinal cord [1,2,3,4,5,6].
Recent progress in medicine, biology and biomaterials engineering in neurosurgery, biomaterial development, cell culture and tissue engineering has allowed for new therapies in SCI. This has contributed to the possibility of healing traumatic SCI and preventing further neurodegeneration [5,6,7,8,9,10,11,12,13,14,15,16,17,18,19]. It remains a severe clinical challenge to effectively treat SCI due to the poor regenerative capacity and complex anatomy of the spinal cord. Several biomaterials that act as scaffolds for axonal growth, cells and neurotrophic factors have become excellent candidates to support the regeneration of the spinal cord.
Recent review articles in new treatments for SCI have mentioned the possibility of clinical applications and the progression of a new regenerative therapy for SCI, including the most recent preclinical results and clinical trials [5,6,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]. However, there have been few review articles in SCI that have mentioned the status of the use of recent biomaterial scaffolds for regenerative therapy and summarized the strategies with simplified results data. In this context, therefore, particular attention has been drawn to biomaterials and nanotechnology-enabled products for the controlled delivery and sustained release of various moieties, including drugs, bioactive molecules and cells [5].
Aiming to set a framework for future clinical use, we briefly describe the most recent developments in biomaterial scaffolds for SCI treatment including combination therapy with cell-seeded materials or innovative drug delivery systems. This review article focuses on previously published biomaterial scaffolds applied to encourage spinal cord regeneration following SCI and summarizes the most recent findings from preclinical and clinical studies using biomaterial scaffolds and other combinatory therapy to treat SCI (Figure 2). An overview of SCI is provided, and the current aspects of clinical biomaterial scaffolds therapy are discussed. First, barriers to regeneration and the pathophysiology of SCI are described. Then, the several categories of biomaterial scaffolds applied in regeneration therapy for SCI are compared. We also review and discuss the current concepts of biomaterial scaffolds in combinatory treatment for SCI and chronic SCI. In the last section, we describe the use of biomaterial scaffolds in ongoing clinical trials for SCI.

2. Barriers to Regeneration and the Pathophysiology of SCI (Figure 1)

The regeneration of the adult mammalian central nervous system (CNS) and spinal cord is difficult due to its limited plasticity [1,6,14,15,16]. Cavitation occurs in the epicenter of a CNS lesion, with this becoming surrounded by connective scar tissue containing cerebrospinal fluid. Reactive astrocytes transform into scar-forming astrocytes that slow the crossing of regenerating axons into the lesion. Certain inflammatory immune cells also remain around the lesion epicenter at the site of the SCI [1,6,14,15,16,17,18,19]. Following SCI, astrocytes are activated, and they proliferate and migrate to the perilesional region to form processes in a dense interwoven network, depositing chondroitin sulfate proteoglycans (CSPGs) into the extracellular matrix (ECM). Dystrophic axons surround the epicenter of the injury and are trapped in the dense meshwork of scar tissue [15,17]. Biomaterial scaffolds that generate specific microenvironmental cues in a three-dimensional (3D), controlled fashion to enhance the survival, infiltration and differentiation of cells [18] are used for spinal cord regeneration following injury.

3. Systematic Review of Biomaterial Scaffolds Applied for SCI

Below, we review the biomaterial scaffolds applied in regeneration therapy for SCI from selected articles following our literature search.

3.1. Literature Search and Inclusion Criteria

In conducting our systematic review, we followed the guidelines of PRISMA (Preferred Reporting Items for Systematic Re-views and Meta-Analyses) (https://prisma-statement.org/, accessed on 1 April 2022). From the PubMed/MEDLINE database, we initially identified relevant articles published up until 30 April 2022 that met the search terms “Spinal cord injury”, and [“Biomaterial”, or “Scaffold”]. After reviewing all of the articles’ titles, we chose titles relevant to our review. Articles not written in English were excluded. After reviewing the abstracts of these titles, we excluded those articles with unrelated titles. Then, following a review of the reference lists in the remaining articles, we identified additional relevant publications and added them. Finally, we performed a full-text review of these articles, and those without a full text available or that were in vitro studies were excluded.
The criteria for article selection were: (1) biomaterial scaffolds were used for SCI or the spinal cord transection model; (2) the treatment outcome was described in detail; and (3) the articles were written in English. In this review of biomaterial scaffolds, we focused on the treatment efficacy of biomaterial scaffolds used in regeneration therapy for SCI in in vivo studies.

3.2. Study Selection

Our database search identified 412 potential articles. A review of the titles and a removal of duplicates resulted in the exclusion of 110 articles, leaving 302 articles for abstract and full-text review, after which 140 articles were excluded. The reasons for study exclusion were: (1) review articles; (2) only protocol papers; (3) treatment outcomes were not described in detail; (4) not suitable after discussion; and (5) not blind studies. After the inclusion of 15 additional relevant publications, 168 studies met the criteria for review. The search flow diagram is depicted in Figure 3. Two reviewers (H.S. and Y.I.) independently screened the titles and abstracts of the studies identified by the search strategy to determine their potential relevance. The full texts of these potentially relevant studies were retrieved, and these same reviewers evaluated them for eligibility. Disagreements were resolved via consensus, and a third independent reviewer (T.S.) resolved any disputes if consensus could not be reached.

3.3. Assessment of Quality and Risk of Bias

Two review authors (H.S. and M.F.) independently assessed the studies for risk for bias using the Cochrane Back Review Group “risk of bias” tool, and a third reviewer (H.I.) and another author (N.N.) helped to resolve any disagreements. All included studies were in vivo animal studies. Therefore, there was no risk of bias in regard to the diagnostic criteria, validity and reliability of the measurements, and no studies had selection bias. In addition, the number of analyzed animals was statistically acceptable in all studies, and they reported the random selection of the control and the scaffold treatment groups.
As shown in tables 168 articles were selected in the present review.

4. Categories of Biomaterial Scaffolds Applied in Regeneration Therapy for SCI

The biomaterial scaffolds used in spinal cord regeneration can be classified according to the required structure and physical and biological properties of the prospective tissue construct applied in SCI. The categories of the biomaterial scaffolds used in spinal cord regeneration include hydrogels, biodegradable scaffolds, the use of micro/nanofibers as instructive biomaterials and drug-delivering biomaterials [29,30,31,32].

4.1. Hydrogels

Hydrogels are one of the most appealing and frequently engineered scaffolds. They are made up of 3D cross-linked biocompatible polymeric macroporous networks that supply the permissive microenvironment and guidance cues necessary for axonal growth [7,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. The hydrogel scaffolds used alone in studies applied for SCI treatment are shown in Table 1 [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. Hydrogels are hydrated networks that mimic the ECM of soft tissues [30,31]. Natural hydrogels usually contain fibrillar proteins within a hydrated glycosaminoglycan network that can enhance cell adhesion and migration in the lesion site. The natural polymers used for nerve tissue engineering include agarose, alginate, chitosan, collagen, fibrin, fibronectin, hyaluronic acid (HA) and Matrigel™ [30,31]. Natural polymers deliver excellent biomimicking, but synthetic hydrogels have also attracted attention because they can potentially control their rate of degradation and for their mechanical properties [30,31].
We reviewed the effects of hydrogel scaffolds on pathophysiologiocal events and motor functional recovery (Table 1) [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. Several types of hydrogels have been reported to date [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. Biopolymer-based hydrogel scaffolds are categorized into natural polymers, synthetic polymers and self-assembling peptides according to the origin of the biomaterial used [7,63]. Twenty-nine articles revealed axonal growth into an implanted biomaterial scaffold [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,60,61,62], and thirteen papers showed motor functional recovery following scaffold implant in in vivo studies [33,36,37,38,43,48,50,53,56,57,58,60]. Several articles revealed an anti-inflammatory effect [38,46,48,50,54,59] and angiogenesis [45,54,55,56,57,58] following the implantation of the biomaterial scaffold in the spinal cord (Table 1).

4.2. Biodegradable Scaffolds

The biodegradable polymers currently used in devices approved by the US Food and Drug Administration provide attractive building blocks for synthetic tissue scaffolds because their biocompatibility has already been established and the regulatory approval process is simple. The biodegradable scaffolds used to treat SCI can be combined with hydrogels. Among the most widely used biodegradable polymers are hydrophobic polyesters such as poly (lactic acid) (PLA), poly (lacticco-glycolic acid) (PLGA) and poly (ε-caprolactone) (PCL). These polymers have been used in sutures and resorbable orthopedic fixation devices because their synthetic fibers provide good mechanical properties and adjustability [53,54]. PLA is a biocompatible lactic acid polymer. The neatly arranged PLA microfibers in transplants promoted the regeneration of CNS tissues [64]. As a product of the reaction between PGA and PLA, which are biodegradable and synthetic polymers, PLGA co-polymer scaffolds show good porosity, hydrophilicity and biodegradability and are usable as drug carriers. One drug delivery device takes the form of a PLGA-based nerve conduit used to control the local delivery of nerve growth factor (NGF) and is applied at the site of the peripheral nerve gap injury [64]. Biocompatible and biodegradable aliphatic polyester make up PCL scaffolds, and this polyester has been used widely in many biomedical applications including bioactive drug delivery for spinal cord regeneration. Other important biomaterials used in SCI include chitosan and gelatin [64]. These are frequently implanted surgically into lesions and are synthesized via electrospinning techniques to decrease organic solvent use [64]. QL6, a biodegradable peptide which self-assembles into nanofiber scaffolds when injected into the spinal cord cavity, has been shown to reduce apoptosis, inflammation and astrogliosis, leading to electrophysiological and behavioral improvements [7,65]. Furthermore, when co-transplanted with NPCs, QL6 enhanced graft survival and promoted differentiation towards neuronal and oligodendroglial cell fates [7,65]. In another type of biodegradable scaffold, functional sequence SIKVAV-modified PA hydrogels implanted into a rat model of SCI improved histological and functional recovery [66].
We reviewed the effects of biodegradable scaffolds on pathophysiologiocal events and motor functional recovery when applied for SCI treatment (Table 2) [66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86]. Most articles revealed axonal growth into implanted biodegradable scaffolds [66,67,68,69,70,71,72,73,74,75,77,79,81,82,83,84,85,86]. Seven papers showed motor functional recovery following scaffold implantation in in vivo studies [69,71,79,81,83,84,85]. Several articles revealed an anti-inflammatory effect [81,84,85] and angiogenesis [66,71,72,82,84,85] following the implantation of the biodegradable scaffold in the spinal cord (Table 2).

4.3. Nano- and Micro-Scale Scaffolds as Instructive Biomaterials for SCI

The recent development of various nanomaterials is offering promising new ways to treat SCI by crossing the blood–spinal cord barrier to deliver therapeutics. Several articles revealed the development of nanomaterials that can modulate inflammatory signals, target inhibitory factors within a lesion and promote axonal regeneration following SCI [87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104].
Experimental models for SCI treatment are increasingly being used to study nanoparticles. The extremely diverse composition of nanoparticles includes polymers, metals and metal oxides, silica and biological molecules [87]. The biocompatibility of polymeric nanoparticles has allowed them to become the most extensively used means of delivering drugs to the spinal cord. Unlike with drugs, topographical cues in the implanted scaffolds at the lesion site can physically guide the extension of new axons [87,88,89,90,91]. The electrospinning of nanofibers is advantageous because it permits the production of highly porous 3D scaffolds with a large surface area that aids in cell adhesion [87]. Spontaneous self-assembling peptides can also form nanofibrous hydrogels that are composed of natural amino acid sequences, rendering them nonimmunogenic, nontoxic and biodegradable [86,87]. Self-assembling peptides have an additional advantage in that they can undergo gelation in physiological conditions, and their morphology mimics in vivo ECM [7,91]. The ionic complementarity of many common self-assembling peptides allows them to form nanofibrous structures. Several articles reported using other materials for nanoscale scaffolds [87,88,89]. Because of their size, which closely mimics that of ECM proteins, and their high surface area, carbon nanotube nanostructures have shown promising effects in neural regeneration applications. Electrospinning produces micro- and nanofibers that can simulate collagen fibers in the ECM [88]. RADA16-I hydrogels were used in an experimental SCI model, which proved that self-assembling peptide hydrogels could promote recovery from SCI [91]. Further development produced functionalized RADA16-I hydrogels with a bone marrow-homing motif (BMHP1) [91,99]. These researchers inserted a 4-glycine-spacer into the hydrogels to facilitate scaffold stability and expose more bi motifs. Their results showed that RADA16-I hydrogels can increase cell infiltration, basement membrane deposition and axon regeneration in SCI [104].
These kinds of nanoscale scaffolds and nanofibers were mainly used for drug delivery systems (DDSs) (refer to Section 5). Therefore, there were only a small number of studies on nanoscale scaffolds applied to SCI treatment [89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104]. We reviewed the effects of nanoscale or microscale biomaterial scaffolds on pathophysiological events and motor functional recovery (Table 3) [89,90,91,92,93,94,95,96,97,98,99,100,101,102,103].

5. Biomaterial Scaffolds in Combinatory Treatment Used for DDSs in SCI Treatment

Hydrogels and biodegradable and nanomaterial scaffolds were also widely used for DDSs as a combinatory treatment for SCI. Scaffolds provide a surrounding 3D environment that promotes the in vivo adhesion, migration and differentiation of cells [104]. In SCIs, the scaffolds, as a matrix for cell, drug and other bioactive molecule delivery, bridge the SC lesion cavity to structurally and chemically support axonal regrowth and stimulate the regeneration of host tissue [104]. Several concepts of SCI treatment using biomaterial scaffolds containing basic fibroblast growth factor, methylprednisolone, calcium responsive composite, neurotrophic factors, anti-Nogo and anti-inflammatory agents have been reported [105,106,107,108,109,110,111]. Strategies for SCI repair are still limited in part by poor drug delivery techniques. However, several ideal DDSs using degradable/nondegradable biomaterial scaffolds have been developed that can provide the localized release of growth factors or other neuroprotective agents from an injectable gel form [112,113,114]. We reviewed the studies of biomaterial scaffolds used in combinatory treatment as DDSs for SCI that are shown in Table 4 [105,106,108,109,110,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130].
Gelatin hydrogel containing basic fibroblast growth factor that was injected into a rat model of SCI showed better performance in relieving mechanical allodynia [105]. Biomaterials containing methylprednisolone can also enhance axonal regeneration and reduce inflammation [106]. The exceptional ability of alginate/chitosan/genipin hydrogels, which show a high sensitivity to Ca2+ composites, to regulate astrocyte behavior and prevent Ca2+-related secondary neuron damage during acute SCI was shown in an in vitro study [107]. A significant therapeutic role was shown for the local delivery of constitutively active Rho GTPases, Cdc42 and Rac1 with the microtubule-mediated slow release of brain-derived neurotrophic factor (BDNF) in overcoming CSPG-mediated failure of regeneration following SCI [108]. A previous article reported on HA hydrogels that were developed to blend with the anti-Nogo receptor antibody (antiNgR). Hydrogel combinations with PLGA microspheres containing BDNF and vascular endothelial growth factor (VEGF) were also reported [109]. Following the implantation of a composite modified by binding with an antiNgR and further mixing with PLGA microspheres containing BDNF and VEGF into an injured area created by the dorsal hemisection of the spinal cord at T9–10 in rats, favorable effects were observed that indicated the promotion of spinal repair, including the integration of the implants with host tissue, the inhibition of inflammation and gliosis [99]. The implantation of bFGF combined with hydroxyl ethyl methacrylate [2-(methacryloyloxy) ethyl] trimethylammonium chloride (HEMA-MOETACL) hydrogels resulted in the promotion of nerve tissue regeneration and functional recovery using hydrogels in a SCI model [110]. These results also suggest the importance of the proper matching of the functional sequence and hydrogels in the synthesis of functional hydrogels. The combination of collagen–laminin scaffolds with 5-NOT treatment also promoted axonal regrowth at the site of SCI as indicated by the expression of NF200 and monoaminergic and glutamatergic reinnervation [113].
Several other combinatory approaches were reported that combined biomaterial scaffolds with rehabilitation and the release from the scaffolds of neurotrophin 3 factor (NT-3), Nogo-66 receptor antibody, ibuprofen/triiodothyronine, sonic hedgehog/retinoic acid, dibutyryl cyclic adenosine monophosphate and rho-A-kinase inhibitor [109,114,115,116,117,118,119,120,121,122,123,124,125]. Other scaffolds, such as silk fibroin combined with neurotrophic factors [125,126], fibrin scaffolds containing growth factors [127] and the polycistronic delivery of IL-10 and NT-3 [128], showed desirable therapeutic potential in terms of SCI treatment. These therapies promoted the differentiation, proliferation and viability of transplanted cells.
The effects of biomaterial scaffolds in combinatory treatments as DDSs applied for SCI treatment on pathophysiological events and motor functional recovery data are summarized in Table 4.

6. Biomaterial Scaffolds in Combinatory Treatment with Cell Therapy for SCI

As an appealing therapeutic approach for SCI, cell therapy can provide significant neuroprotection, the recovery of functionaility through cell replacement, trophic support and the modulation of immune factors [18,131], and, thus, clinical trials have also been started in humans [132,133]. As mentioned above, we noted the use of biomaterials for SCI repair because of the structural or active growth support they provide to damaged axons. Furthermore, biomaterials have the ability to function as cell delivery platforms for cells and therapeutic molecules and as a local depot for sustained drug release. Both cell regeneration and tissue reconstruction can be achieved when these two therapeutic methods are combined. By following the basic operating principle of this modality, i.e., the combination of exogenous cells and scaffolds to form live scaffolds, we can expect the synergic effects of stem cells and scaffolds to occur. These live scaffolds can be implanted into animals through injection or surgical implantation without side effects [104]. We reviewed the preclinical studies using biomaterial scaffolds in combination with cell therapy, so called multipotent stem cells, for the treatment of SCI. The combinatory treatments with cell therapy are summarized in Table 5 [36,117,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227].

6.1. Exogenous Neural Stem/Progenitor Cells and Biomaterial Scaffolds

Therapies using exogenous neural stem/progenitor cells (NPCs) show particular promise because these cells can potentially differentiate into all three neuroglial lineages—neurons, astrocytes and oligodendrocytes—to regenerate neural circuits, remyelinate denuded axons and provide trophic support to endogenous cells [15,18,19,131]. However, the transplantation of NPCs, especially in the chronic phase, showed several issues regarding tissue regeneration in terms of the survival rate of NPCs and insufficient integration with injured spinal cord [15,18]. Many researchers have tried novel combinatorial treatments with biomaterial scaffolds and NPCs, and several articles have reported the expected synergic effects of these grafts [15,65]. Over the previous decade, when NSCs were delivered via a supporting scaffold matrix, significant outcomes regarding functional recovery were consistently observed in the preclinical stage [6,7,9,19,65], but these favorable results have yet to be translated into clinical use. In the meantime, clinical developments that affect the safety and feasibility of implantable biomaterials for CNS repair are currently underway. The safety and feasibility of the transplantation of the NeuroRegen implantable collagen scaffold in completely chronic patients with SCI has been reported, although the sample size in these studies is small [132,133].
NPCs derived from induced pluripotent stem cells (iPSCs), embryonic stem cells or brain or spinal cord within biomaterial scaffolds are also being used for the treatment of SCI [36,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177] (Table 5). SCI rats receiving the transplantation of NPCs in Matrigel showed improvements in behavioral recovery and the expression levels of neuronal and reactive astrocyte markers [162]. A fabricated biodegradable hybrid inorganic scaffold comprised of biodegradable MnO2 nanosheets enhanced the attachment and differentiation of iPSC-derived NSCs in the site of SCI [134]. Fibrin scaffolds and stem cell therapy designed to immobilize cells and release growth factors (NT3, glial-derived neurotrophic factor [GDNF] and platelet-derived growth factor-A [PDGF]) from fibrin achieved better recovery from SCI [158,159]. NPCs used with self-assembling peptide QL6 decreased the formation of cystic cavities and inflammation and enhanced synaptic connections through a reduction in astrogliosis and CSPG, thus improving forelimb functionality in an SCI model of cervical injury [7,65]. Laminin-coated hydrogel enhanced iPSC-NPC viability and promoted host axon and astrocyte growth in lesion sites [152]. Another article reported the manufacture of NPCs biased toward an oligodendrogenic fate and the upgrading of the ChABC delivery system via a crosslinked methylcellulose biomaterial. This combinatorial therapy resulted in the promotion of oligodendrocyte differentiation, remyelination and synaptic connectivity [165]. A linearly ordered collagen scaffold modified with N-cadherin promoted the migration and differentiation of endogenous neural/progenitor stem cells, which produced a desirable therapeutic effect in rats following SCI [36]. One treatment showing great potential for SCI treatment was the combination of a collagen microchannel scaffold and paclitaxel liposome, which induced the neuronal differentiation of NSCs and neuron and axon growth [144]. A different group reported the benefits of combining NPCs and K2(QL)6K2 (QL6), an aqueous self-assembling peptide that aggregates into a stable nanofiber gel due to multiple non-covalent interactions [7,103]. In a study exploring the modification of a scaffold with PDGF-A to induce oligodendrocyte differentiation, NPCs cultured in a hydrogel blend of hyaluronan and methylcellulose (HAMC) modified with PDGF-A showed improved survival and the higher differentiation of cells into oligodendrocytes. SCI rats transplanted with NPCs cultured in this hydrogel blend showed reduced cavitation, improved graft survival with increased differentiation of oligodendrocytes and improved behavioral recovery [170]. These researchers further modified the HAMC-PDGF-A scaffold with arginine-glycine-aspartic acid (RGD) peptide to improve the engraftment and survival of human iPSC-derived oligodendrocyte precursor cells (OPCs). iPS cell-derived OPCs transplanted in HAMC-RGD/PDGF-A had higher rates of survival and engraftment than iPS cell-derived OPCs transplanted with media did [171].

6.2. Nanoscaffolds and Stem Cell Grafts

Neuroinflammatory agents such as metalloproteinase and neurotoxic cytokines that are secreted after CNS injuries can lead to a reduction in the neuroinhibitory microenvironment in the region of injury [177]. Drug-loaded 3D nanoscaffolds designed to reduce neuroinflammatory agents were fabricated using a layer-by-layer method in which chitosan polymer functionalized manganese oxide nanosheets for fabrication into a 3D esoporous structure. Methylprednisolone and laminin were also added as cell-adherent ECM ligands to the fabricated scaffold, and its effects on SCI treatment were evaluated with iPSCs. This study confirmed functional recovery and axonal growth due to stem cell differentiation and the suppression of fibrotic scar fabrication in an in vivo model of SCI [177,178]. By mimicking the ECM, the fibrous structure of the nanofibrous scaffolds provided an ideal platform for the attachment, proliferation and differentiation of stem cells [178]. The capability of multichannel nanofibrous scaffolds using poly-L-lysine integrated NT-3 to promote the recruitment and differentiation of endogenous NPCs facilitated synapse formation and enhanced locomotor recovery, thus promoting the treatment of SCI [166].

6.3. Mescenchymal Stem Cells (MSCs) and Biomaterial Scaffolds: Bone Marrow MSCs, Umbilical MSCs, Wharton’s Jelly-Derived MSCs and Adipose-Derived MSCs

MSCs are a type of stem cell present in adults that can differentiate into mesodermal-derived tissues such as bone, cartilage, blood vessels and cardiomyocytes. MSCs used for clinical purposes are derived from tissues such as bone marrow, umbilical cord and cord blood and fat. MSCs have important biological activities for tissue repair, such as anti-inflammatory effects, growth factor secretion and the promotion of angiogenesis in addition to having a low risk of tumor formation. Moreover, MSCs exhibit remarkable autocrine and paracrine activity. MSCs can secrete various soluble molecules that exert anti-inflammatory potential, including tumor necrosis factor (TNF)-β1, interleukin (IL)-13, IL-18 binding protein, ciliary neurotrophic factor (CNTF), NT-3, IL-10-, IL-12p70, IL-17E and IL-27 [179]. Furthermore, the release of pro-inflammatory cytokines such as interferon, TNF and IL-10 can also be inhibited by MSCs to modulate cytokine production in the host. These cells also produce a wide variety of growth-promoting molecules, including BDNF, CNTF, GDNF, leukemia inhibitory factor, NGF and neurotrophin 3 (NT-3) and ECM proteins such as laminin, fibronectin and collagen I/III and IV [176]. One essential method used by MSCs to secrete biological factors is through extracellular vesicles, which include microvesicles and exosomes. [179]. The co-transplantation of biomaterial and MSCs that have been manipulated or genetically edited to express certain proteins causes neuroprotective and anti-inflammatory effects that induce anti-inflammatory mechanisms [180]. The transplantation of biomaterial-supported MSCs lessens fibrosis during the early process of secondary SCI and further attenuates secondary glial scarring [181]. Biomaterial-supported MSCs that were transplanted into the damaged region subsequently prevented the accumulation of CSPGs, which make up the glial scar, and significantly promoted the myelination of axon fibers and synapse formation [181]. MSCs can cooperate with biomaterials to support the growth of stem cells and endogenous neuronal cells by bridging the gap. A nanofibrous scaffold of polypyrrole/polylactic acid was also used as a platform to deliver bone marrow mesenchymal stem cells (BMSCs) to the site of SCI. BMSCs are beneficial cells with the ability to differentiate into different neural cell types and appear to be proper candidates for replacing damaged cells in SCI. Furthermore, these cells secrete neurotrophic factors to protect the injured spinal cord [182]. The application of this formulation promoted myelination and axon regeneration, enhanced the microenvironment at the site of injury and synergically reduced neuronal apoptosis at the injury site in the spinal cord [182].
The combination of Matrigel and neural-induced adipose-derived MSCs reduced fibrosis from secondary injury processes and improved neuronal regeneration [182]. According to a behavioral and electrophysiological analysis, 3D-printed collagen/silk fibrin scaffolds carrying umbilical secretomes of MSCs improved hindlimb locomotor functionality [183]. Wharton’s jelly-derived MSCs applied with integrin-binding peptide RGD bridged the lesion cavity, supported vascularization, upregulated related gene expressions and increased axonal sprouting into the lesion [184]. The transplantation of human umbilical cord MSCs seeded in collagen scaffolds also reduced scar formation and promoted functional recovery in chronic SCI [183,185].
The combination therapies of biomaterial scaffolds and MCSs for SCI are summarized in Table 5 [181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,217]. Several articles used combinations with cells other than NPCs, i.e., Schwann cells (SCs) and/or olfactory ensheathing cells, to support the survival, integration and migration of grafted cells [163,218,219,224].

6.4. Schwann Cells, Olfactory Ensheathing Cells, Astrocytes and Other Cell Grafts and Biomaterial Scaffolds

SCs are neuroglial cells that drive axon regeneration and myelination in the peripheral nervous system, but they also perform an analogous function when transplanted into the spinal cord. Furthermore, SCs can be isolated from a patient’s own nerves and expanded in vitro prior to implantation, making them an exceptional cell type for autotransplantation therapy in SCI [216]. Numerous preclinical studies have established the functionality of SCs in transplantation [211]. In this section, we review the studies on the combination of biomaterial scaffolds with SCs [210,211,212,213,214,215,216,217,218,219,220,221,222,223,224] shown in Table 5.
One study showed that long-distance regeneration could occur from CNS neurons that project through a scaffold construct into distal tissue implanted with biodegradable PLGA scaffolds loaded with SCs [154]. Other studies found that a poly-b-hydroxybutyrate scaffold, positively charged oligo[poly(ethylene glycol) fumarate] (OPF+) or resorbable poly(-hydroxyacid) guidance channels containing SCs promoted the attachment, proliferation and survival of grafted cells and supported marked axonal regeneration within the graft [210,211,212,213,214,215,216,217,218,219].
The efficacy of other combinatory cell sources on SCI treatment was also reported. The transplantation into an SCI rat model of dental pulp stem cells combined with chitosan scaffolds resulted in the marked recovery of hind limb locomotor functions by increasing the levels of BDNF, GDNF, beta-NGF and NT-3 [220]. The support of the spinal cord structure and induction of cell/tissue polarity were also achieved by the injection of dental follicle cells combined with aligned PCL/PLGA electrospun material [221]. In glial scars, astrocytes have been shown to be important for spontaneous recovery from SCI. One paper reported the effect of implanting HA hydrogels containing ECM harvested from embryonic stem cell-derived astrocytes on histologic outcomes following SCI in rats. Protoplasmic embryonic stem-derived astrocyte ECM also showed the potential to treat SCI injury [222]. PLGA complexes inoculated with olfactory ensheathing cells improved the recovery of locomotor functionality in rat models with transected SCI, most likely because these complexes are conducive to a relatively benevolent microenvironment, offer nerve-protective effects and have the ability to enhance remyelination via the promotion of cell differentiation and the inhibition of astrocyte formation [223,224].
Several articles reported on the co-transplantation of several stem cell types and scaffolds [151,153,155,163,218,219,224]. The effect provided by the co-transplantation of NPCs, SCs and PLGA resulted in better behavioral recovery than that from transplantation with NPCs/PLGA alone [151,153,155]. Axonal regeneration and functional recovery in rat SCI were improved after use of a multichannel polymer scaffold seeded with activated SCs and BMSCs rather than by single treatment with each cell type [219].

7. Biomaterial Scaffolds in Regeneration Therapy for Chronic SCI

Several combinatory treatments for chronic SCI using stem cells and biomaterial scaffolds were recently reported clinically and in rodent models [36,132,134,202,206,225,226,227,228,229,230,231,232,233,234,235,236]. The creation of an artificial scaffold that mimics the ECM and supports nervous system regeneration remains one of the greatest challenges in regeneration following chronic SCI.
One effective measure to repair chronic SCI is the removal of scar tissue combined with biomaterial implantation [44]. One article revealed that following scar tissue removal in chronic SCI, the implantation of a Taxol-modified linear-ordered collagen scaffold (LOCS + Taxol) could promote axonal regeneration, neurogenesis and electrophysiological and functional recovery [44]. Pivotal features of neural repair were also shown following treatment with reduced graphene oxide scaffolds at 4 months after SCI [224]. These results indicate that even if a patient is in the chronic phase of SCI, the potential for axonal regeneration, neurogenesis and functional recovery are still preserved at the site of the SCI.
The use of other scaffolds to bridge defects was reported in experimental models of chronic SCI [172,225,227,229,230,235,236]. Following the removal of scarring, anisotropic alginate hydrogel scaffolds promoted axonal growth across chronic transections of the spinal cord [234]. Engraftment with this scaffold significantly improved electrophysiological conductivity and locomotor functionality. Scar formation was reduced and functional recovery in chronic SCI was promoted following the transplantation of human umbilical cord-derived MSCs seeded in collagen scaffolds [206,225]. Other articles revealed the efficacy of laminin-coated pHEMA-MOETACl hydrogel [152], HPMA-RGD hydrogels [67] and chimeric self-assembling nanofiber [227,232], but these were combined with iPSC-derived NPCs or MSCs. The treatment of chronic SCI with 3D-aligned nanofiber-hydrogel scaffolds [43,92,229], self-assembling scaffolds, Taxol-modified collagen scaffolds [44], graphene oxide scaffolds [99,235] and nanostructured composite scaffolds [229] were also reported. These articles showed that it is possible to recreate an anatomical, structural and histological framework that can allow for replacement of large hollow tissue gaps in chronically injured spinal cord and encourage axonal regeneration and neurological recovery.
As indicated by many researchers, a multi-disciplinary approach is required to solve the problem of repairing chronic SCI. From this point of view, combinatory treatment using stem cells and biological scaffolds will be an important approach in the treatment of chronic SCI in the future [18,19].

8. Biomaterial Scaffolds in Clinical Trials for SCI

In this section, we review the published and ongoing clinical trials of biomaterial scaffolds for SCI (Table 6) (https://www.clinicaltrials.gov/, accessed on 1 August 2022).
One group reported the results of the NeuroRegen clinical trial using the same protocol as that described in Section 6 [132,133,226,233,236,237]. They revealed that the following primary efficacy outcomes of combinatory therapy with BMSCs or MSCs were observed in some patients with chronic SCI: an expansion of the sensation level and motor-evoked potential (MEP)-responsive area, increased activity in the fingers, an enhancement in trunk stability, the return of the sensation of defecation and the recovery of autonomic neural functionality [132,133,226].
Another group provided the result of bridging defects in chronic SCI in a clinical trial using a combination of peripheral nerve grafts and a chitosan–laminin scaffold. Treatment with this combination enhanced regeneration through co-transplantation with bone-marrow-derived MSCs [237]. The grade on the impairment scale of the American Spinal Impairment Association (ASIA) improved from A to C in 12 patients and from A to B in 2 patients [226].
In the phase 2 NCT02688049 clinical study which begun in January 2016, patients with chronic SCI (ASIA grade A) are receiving a NeuroRegen scaffold transplanted with 10 million NSCs after localized scarring is cleared, and after the surgery patients undergo comprehensive rehabilitation combined with psychological and nutritional measures. Ongoing clinical trials of the NeuroRegen Scaffold with the transplantation of BMSCs or MSCs are also being performed in phase 1 and 2 (NCT02352077, NCT02688062) trials. Other clinical studies are ongoing using collagen scaffolds, the RMx Biomatrix or the transplantation of the poly(lactic-co-glycolic acid)-b-poly(L-lysine) scaffold (NCT02510365, NCT03966794, NCT02326662, NCT03762655, NCT02138110), as shown in Table 6.

9. Conclusions and Outlook

In this review, we summarized the most recent insights of the 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 separately summarized the preclinical experimental results for hydrogels, biodegradable scaffolds, nano-/microscale scaffolds, biomaterial scaffolds in combinatory treatment used for DDSs, combinatory with cell therapy and regeneration therapy for chronic SCI. In addition, in the last section, we also reviewed ongoing and the most recently completed clinical trials using biomaterial scaffolds for SCI. Presently, a number of clinical and experimental studies have reported positive results showing motor functional improvement, anti-inflammation, scar/cavity reduction, axon growth and angiogenesis promotion in SCI with the use of biomaterial scaffold grafts. Although some inherent limitations still exist in performing human SCI trials, in that animal experiments cannot be directly applied to humans, much basic research and many clinical trials of biomaterial scaffold therapy have already been performed that show promising results. This database could serve as a benchmark for progress in future clinical trials for SCI with biomaterial scaffolds. Nevertheless, we strongly believe that in the near future, biomaterial scaffolds will deliver the radical treatment required to treat patients with SCI.

Author Contributions

H.S., Y.I. and T.S. designed the outline of the review; H.S., H.I., M.F. and N.N. drafted the manuscript; T.S. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the MHLW FG Program, grant number [JPMH22FG2001].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fehlings, M.G.; Martin, A.R.; Tetreault, L.A.; Aarabi, B.; Anderson, P.; Arnold, P.M.; Brodke, D.; Burns, A.; Chiba, K.; Dettori, J.R.; et al. A clinical practice guideline for the management of patients with acute spinal cord injury: Recommendations on the role of baseline magnetic resonance imaging in clinical decision making and outcome prediction. Glob. Spine J. 2017, 7, 221S–230S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Fehlings, M.G.; Kim, K.D.; Aarabi, B.; Rizzo, M.; Bond, L.M.; McKerracher, L.; Vaccaro, A.R.; Okonkwo, D.O. Rho Inhibitor VX-210 in Acute Traumatic Subaxial Cervical Spinal Cord Injury: Design of the SPinal Cord Injury Rho INhibition InvestiGation (SPRING) Clinical Trial. J. Neurotrauma 2018, 35, 1049–1056. [Google Scholar] [CrossRef] [PubMed]
  3. Singh, A.; Tetreault, L.; Kalsi-Ryan, S.; Nouri, A.; Fehlings, M.G. Global prevalence and incidence of traumatic spinal cord injury. Clin. Epidemiol. 2014, 6, 309–331. [Google Scholar] [CrossRef] [Green Version]
  4. Spinal Cord Injury (SCI) 2016 Facts and Figures at a Glance. J. Spinal Cord Med. 2016, 39, 493–494. [CrossRef] [Green Version]
  5. Costăchescu, B.; Niculescu, A.-G.; Dabija, M.G.; Teleanu, R.I.; Grumezescu, A.M.; Eva, L. Novel Strategies for Spinal Cord Regeneration. Int. J. Mol. Sci. 2022, 23, 4552. [Google Scholar] [CrossRef]
  6. Ahuja, C.S.; Fehlings, M. Concise Review: Bridging the Gap: Novel Neuroregenerative and Neuroprotective Strategies in Spinal Cord Injury. Stem Cells Transl. Med. 2016, 5, 914–924. [Google Scholar] [CrossRef] [Green Version]
  7. Zweckberger, K.; Ahuja, C.S.; Liu, Y.; Wang, J.; Fehlings, M.G. Self-assembling peptides optimize the post-traumatic milieu and synergistically enhance the effects of neural stem cell therapy after cervical spinal cord injury. Acta Biomater. 2016, 42, 77–89. [Google Scholar] [CrossRef] [PubMed]
  8. Kawabata, S.; Takano, M.; Numasawa-Kuroiwa, Y.; Itakura, G.; Kobayashi, Y.; Nishiyama, Y.; Sugai, K.; Nishimura, S.; Iwai, H.; Isoda, M.; et al. Grafted Human iPS Cell-Derived Oligodendrocyte Precursor Cells Contribute to Robust Remyelination of Demyelinated Axons after Spinal Cord Injury. Stem Cell Rep. 2016, 6, 1–8. [Google Scholar] [CrossRef] [Green Version]
  9. Lu, P.; Kadoya, K.; Tuszynski, M.H. Axonal growth and connectivity from neural stem cell grafts in models of spinal cord injury. Curr. Opin. Neurobiol. 2014, 27, 103–109. [Google Scholar] [CrossRef]
  10. Lis, A.; Szarek, D.; Laska, J. Strategie inzynierii biomateriałów dla regeneracji rdzenia kregowego: Aktualny stan wiedzy. Polym. Med. 2013, 43, 59–80. [Google Scholar]
  11. Imagama, T.; Ogino, K.; Takemoto, K.; Kato, Y.; Kataoka, H.; Suzuki, H.; Ran, Z.; Setiawan, H.; Fujikura, Y.; Taguchi, T. Regulation of nitric oxide generation by up-regulated arginase I in rat spinal cord injury. J. Clin. Biochem. Nutr. 2012, 51, 68–75. [Google Scholar] [CrossRef] [PubMed]
  12. Murakami, T.; Kanchiku, T.; Suzuki, H.; Imajo, Y.; Yoshida, Y.; Nomura, H.; Cui, D.; Ishikawa, T.; Ikeda, E.; Taguchi, T. Anti-interleukin-6 receptor antibody reduces neuropathic pain following spinal cord injury in mice. Exp. Ther. Med. 2013, 6, 1194–1198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Suzuki, H.; Taguchi, T.; Kato, Y.; Kanchiku, T.; Imagama, T.; Yara, T.; Moriya, A.; Muramatsu, K.; Tanaka, H.; Gondo, T. Transplantation of neurospheres derived from bone marrow stromal cells promotes neurological recovery in rats with spinal cord injury. Med. Mol. Morphol. 2011, 44, 131–138. [Google Scholar] [CrossRef] [PubMed]
  14. Wilcox, J.T.; Satkunendrarajah, K.; Zuccato, J.A.; Nassiri, F.; Fehlings, M.G. Neural Precursor Cell Transplantation Enhances Functional Recovery and Reduces Astrogliosis in Bilateral Compressive/Contusive Cervical Spinal Cord Injury. Stem Cells Transl. Med. 2014, 3, 1148–1159. [Google Scholar] [CrossRef] [PubMed]
  15. Suzuki, H.; Ahuja, C.S.; Salewski, R.P.; Li, L.; Satkunendrarajah, K.; Nagoshi, N.; Shibata, S.; Fehlings, M.G. Neural stem cell mediated recovery is enhanced by Chondroitinase ABC pretreatment in chronic cervical spinal cord injury. PLoS ONE 2017, 12, e0182339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ramon y Cajal, S. Degeneration and Regeneration of the Nervous System; Oxford University Press: London, UK, 1928. [Google Scholar]
  17. Bradbury, E.J.; Moon, L.D.F.; Popat, R.J.; King, V.R.; Bennett, G.S.; Patel, P.N.; Fawcett, J.W.; McMahon, S.B. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature 2002, 416, 636–640. [Google Scholar] [CrossRef]
  18. Suzuki, H.; Imajo, Y.; Funaba, M.; Nishida, N.; Sakamoto, T.; Sakai, T. Current Concepts of Neural Stem/Progenitor Cell Therapy for Chronic Spinal Cord Injury. Front. Cell. Neurosci. 2022, 15, 794692. [Google Scholar] [CrossRef]
  19. Suzuki, H.; Sakai, T. Current Concepts of Stem Cell Therapy for Chronic Spinal Cord Injury. Int. J. Mol. Sci. 2021, 22, 7435. [Google Scholar] [CrossRef]
  20. Ahuja, C.S.; Nori, S.; Tetreault, L.; Wilson, J.; Kwon, B.; Harrop, J.; Choi, D.; Fehlings, M.G. Traumatic Spinal Cord Injury—Repair and Regeneration. Neurosurgery 2017, 80, S9–S22. [Google Scholar] [CrossRef]
  21. Cofano, F.; Boido, M.; Monticelli, M.; Zenga, F.; Ducati, A.; Vercelli, A.; Garbossa, D. Mesenchymal Stem Cells for Spinal Cord Injury: Current Options Limitations, and Future of Cell Therapy. Int. J. Mol. Sci. 2019, 20, 2698. [Google Scholar] [CrossRef] [Green Version]
  22. Hutson, T.H.; Di Giovanni, S. The translational landscape in spinal cord injury: Focus on neuroplasticity and regeneration. Nat. Rev. Neurol. 2019, 15, 732–745. [Google Scholar] [CrossRef] [PubMed]
  23. Badhiwala, J.H.; Wilson, J.R.; Kwon, B.K.; Casha, S.; Fehlings, M.G. A Review of Clinical Trials in Spinal Cord Injury Including Biomarkers. J. Neurotrauma 2018, 35, 1906–1917. [Google Scholar] [CrossRef] [PubMed]
  24. Shinozaki, M.; Nagoshi, N.; Nakamura, M.; Okano, H. Mechanisms of Stem Cell Therapy in Spinal Cord Injuries. Cells 2021, 10, 2676. [Google Scholar] [CrossRef] [PubMed]
  25. Ahuja, C.S.; Mothe, A.; Khazaei, M.; Badhiwala, J.H.; Gilbert, E.A.; van der Kooy, D.; Morshead, C.M.; Tator, C.; Fehlings, M.G. The leading edge: Emerging neuroprotective and neuroregenerative cell-based therapies for spinal cord injury. Stem Cells Transl. Med. 2020, 9, 1509–1530. [Google Scholar] [CrossRef] [PubMed]
  26. Gabel, B.C.; Curtis, E.I.; Marsala, M.; Ciacci, J.D. A Review of Stem Cell Therapy for Spinal Cord Injury: Large Animal Models and the Frontier in Humans. World Neurosurg. 2017, 98, 438–443. [Google Scholar] [CrossRef]
  27. Zipser, C.M.; Cragg, J.J.; Guest, J.D.; Fehlings, M.G.; Jutzeler, C.R.; Anderson, A.J.; Curt, A. Cell-based and stem-cell-based treatments for spinal cord injury: Evidence from clinical trials. Lancet Neurol. 2022, 21, 659–670. [Google Scholar] [CrossRef]
  28. Kiyotake, E.A.; Martin, M.D.; Detamore, M.S. Regenerative rehabilitation with conductive biomaterials for spinal cord injury. Acta Biomater. 2022, 139, 43–64. [Google Scholar] [CrossRef]
  29. Ghane, N.; Beigi, M.-H.; Labbaf, S.; Nasr-Esfahani, M.-H.; Kiani, A. Design of hydrogel-based scaffolds for the treatment of spinal cord injuries. J. Mater. Chem. B 2020, 8, 10712–10738. [Google Scholar] [CrossRef]
  30. Pina, S.; Ribeiro, V.P.; Marques, C.F.; Maia, F.R.; Silva, T.H.; Reis, R.L.; Oliveira, J.M. Scaffolding Strategies for Tissue Engineering and Regenerative Medicine Applications. Materials 2019, 12, 1824. [Google Scholar] [CrossRef] [Green Version]
  31. Estrada, V.; Tekinay, A.; Müller, H.W. Neural ECM mimetics. Prog. Brain Res. 2014, 214, 391–413. [Google Scholar] [CrossRef]
  32. Khaing, Z.Z.; Seidlits, S.K. Hyaluronic acid and neural stem cells: Implications for biomaterial design. J. Mater. Chem. B 2015, 3, 7850–7866. [Google Scholar] [CrossRef] [PubMed]
  33. Sun, Y.; Yang, C.; Zhu, X.; Wang, J.-J.; Liu, X.-Y.; Yang, X.-P.; An, X.-W.; Liang, J.; Dong, H.-J.; Jiang, W.; et al. 3D printing collagen/chitosan scaffold ameliorated axon regeneration and neurological recovery after spinal cord injury. J. Biomed. Mater. Res. Part A 2019, 107, 1898–1908. [Google Scholar] [CrossRef] [PubMed]
  34. Marchand, R.; Woerly, S. Transected spinal cords grafted with in situ self-assembled collagen matrices. Neuroscience 1990, 36, 45–60. [Google Scholar] [CrossRef] [PubMed]
  35. Khan, T.; Dauzvardis, M.; Sayers, S. Carbon filament implants promote axonal growth across the transected rat spinal cord. Brain Res. 1991, 541, 139–145, Erratum in Brain Res. 1991, 546, 360. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, W.; Xu, B.; Xue, W.; Yang, B.; Fan, Y.; Chen, B.; Xiao, Z.; Xue, X.; Sun, Z.; Shu, M.; et al. A functional scaffold to promote the migration and neuronal differentiation of neural stem/progenitor cells for spinal cord injury repair. Biomaterials 2020, 243, 119941. [Google Scholar] [CrossRef]
  37. Fan, C.; Li, X.; Xiao, Z.; Zhao, Y.; Liang, H.; Wang, B.; Han, S.; Li, X.; Xu, B.; Wang, N.; et al. A modified collagen scaffold facilitates endogenous neurogenesis for acute spinal cord injury repair. Acta Biomater. 2017, 51, 304–316. [Google Scholar] [CrossRef]
  38. Yang, B.; Liang, C.; Chen, D.; Cheng, F.; Zhang, Y.; Wang, S.; Shu, J.; Huang, X.; Wang, J.; Xia, K.; et al. A conductive supramolecular hydrogel creates ideal endogenous niches to promote spinal cord injury repair. Bioact. Mater. 2022, 15, 103–119. [Google Scholar] [CrossRef]
  39. Martín-López, E.; Darder, M.; Ruiz-Hitzky, E.; Sampedro, M.N. Agar-based bridges as biocompatible candidates to provide guide cues in spinal cord injury repair. Bio-Med. Mater. Eng. 2013, 23, 405–421. [Google Scholar] [CrossRef]
  40. Gros, T.; Sakamoto, J.S.; Blesch, A.; Havton, L.A.; Tuszynski, M.H. Regeneration of long-tract axons through sites of spinal cord injury using templated agarose scaffolds. Biomaterials 2010, 31, 6719–6729. [Google Scholar] [CrossRef]
  41. Kataoka, K.; Suzuki, Y.; Kitada, M.; Hashimoto, T.; Chou, H.; Bai, H.; Ohta, M.; Wu, S.; Suzuki, K.; Ide, C. Alginate Enhances Elongation of Early Regenerating Axons in Spinal Cord of Young Rats. Tissue Eng. 2004, 10, 493–504. [Google Scholar] [CrossRef]
  42. Prang, P.; Mueller, R.; Eljaouhari, A.; Heckmann, K.; Kunz, W.; Weber, T.; Faber, C.; Vroemen, M.; Bogdahn, U.; Weidner, N. The promotion of oriented axonal regrowth in the injured spinal cord by alginate-based anisotropic capillary hydrogels. Biomaterials 2006, 27, 3560–3569. [Google Scholar] [CrossRef] [PubMed]
  43. Cao, Z.; Yao, S.; Xiong, Y.; Zhang, Z.; Yang, Y.; He, F.; Zhao, H.; Guo, Y.; Wang, G.; Xie, S.; et al. Directional axonal regrowth induced by an aligned fibrin nanofiber hydrogel contributes to improved motor function recovery in canine L2 spinal cord injury. J. Mater. Sci. Mater. Med. 2020, 31, 40. [Google Scholar] [CrossRef] [PubMed]
  44. Yin, W.; Xue, W.; Zhu, H.; Shen, H.; Xiao, Z.; Wu, S.; Zhao, Y.; Cao, Y.; Tan, J.; Li, J.; et al. Scar tissue removal-activated endogenous neural stem cells aid Taxol-modified collagen scaffolds in repairing chronic long-distance transected spinal cord injury. Biomater. Sci. 2021, 9, 4778–4792. [Google Scholar] [CrossRef]
  45. Altinova, H.; Hammes, S.; Palm, M.; Achenbach, P.; Gerardo-Nava, J.; Deumens, R.; Führmann, T.; van Neerven, S.G.; Hermans, E.; Weis, J.; et al. Dense fibroadhesive scarring and poor blood vessel-maturation hamper the integration of implanted collagen scaffolds in an experimental model of spinal cord injury. Biomed. Mater. 2020, 15, 015012. [Google Scholar] [CrossRef] [PubMed]
  46. Gholami, M.; Gilanpour, H.; Sadeghinezhad, J.; Asghari, A. Facile fabrication of an erythropoietin-alginate/chitosan hydrogel and evaluation of its local therapeutic effects on spinal cord injury in rats. DARU J. Pharm. Sci. 2021, 29, 255–265. [Google Scholar] [CrossRef]
  47. Stokols, S.; Tuszynski, M.H. Freeze-dried agarose scaffolds with uniaxial channels stimulate and guide linear axonal growth following spinal cord injury. Biomaterials 2006, 27, 443–451. [Google Scholar] [CrossRef]
  48. Zhang, Z.; Yao, S.; Xie, S.; Wang, X.; Chang, F.; Luo, J.; Wang, J.; Fu, J. Effect of hierarchically aligned fibrin hydrogel in regeneration of spinal cord injury demonstrated by tractography: A pilot study. Sci. Rep. 2017, 7, 40017. [Google Scholar] [CrossRef] [Green Version]
  49. Fukushima, K.; Enomoto, M.; Tomizawa, S.; Takahashi, M.; Wakabayashi, Y.; Itoh, S.; Kuboki, Y.; Shinomiya, K. The axonal regeneration across a honeycomb collagen sponge applied to the transected spinal cord. J. Med. Dent. Sci. 2008, 55, 71–79. [Google Scholar]
  50. Zhao, X.; Wang, H.; Zou, Y.; Xue, W.; Zhuang, Y.; Gu, R.; Shen, H.; Dai, J. Optimized, visible light-induced crosslinkable hybrid gelatin/hyaluronic acid scaffold promotes complete spinal cord injury repair. Biomed. Mater. 2022, 17, 024104. [Google Scholar] [CrossRef]
  51. King, V.R.; Alovskaya, A.; Wei, D.; Brown, R.A.; Priestley, J.V. The use of injectable forms of fibrin and fibronectin to support axonal ingrowth after spinal cord injury. Biomaterials 2010, 31, 4447–4456. [Google Scholar] [CrossRef]
  52. Cheng, H.; Huang, Y.-C.; Chang, P.-T.; Huang, Y.-Y. Laminin-incorporated nerve conduits made by plasma treatment for repairing spinal cord injury. Biochem. Biophys. Res. Commun. 2007, 357, 938–944. [Google Scholar] [CrossRef] [PubMed]
  53. Han, S.; Lee, J.Y.; Heo, E.Y.; Kwon, I.K.; Yune, T.Y.; Youn, I. Implantation of a Matrigel-loaded agarose scaffold promotes functional regeneration of axons after spinal cord injury in rat. Biochem. Biophys. Res. Commun. 2018, 496, 785–791. [Google Scholar] [CrossRef] [PubMed]
  54. Bakshi, A.; Fisher, O.; Dagci, T.; Himes, B.T.; Fischer, I.; Lowman, A. Mechanically engineered hydrogel scaffolds for axonal growth and angiogenesis after transplantation in spinal cord injury. J. Neurosurg. Spine 2004, 1, 322–329. [Google Scholar] [CrossRef]
  55. Zhai, H.; Zhou, J.; Xu, J.; Sun, X.; Xu, Y.; Qiu, X.; Zhang, C.; Wu, Z.; Long, H.; Bai, Y.; et al. Mechanically strengthened hybrid peptide-polyester hydrogel and potential applications in spinal cord injury repair. Biomed. Mater. 2020, 15, 055031. [Google Scholar] [CrossRef] [PubMed]
  56. Hejčl, A.; Růžička, J.; Kekulová, K.; Svobodová, B.; Proks, V.; Macková, H.; Jiránková, K.; Kárová, K.; Urdziková, L.M.; Kubinová, Š.; et al. Modified Methacrylate Hydrogels Improve Tissue Repair after Spinal Cord Injury. Int. J. Mol. Sci. 2018, 19, 2481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Zhang, Q.; Yan, S.; You, R.; Kaplan, D.L.; Liu, Y.; Qu, J.; Li, X.; Li, M.; Wang, X. Multichannel silk protein/laminin grafts for spinal cord injury repair. J. Biomed. Mater. Res. Part A 2016, 104, 3045–3057. [Google Scholar] [CrossRef]
  58. Chai, Y.; Long, Y.; Dong, X.; Liu, K.; Wei, W.; Chen, Y.; Qiu, T.; Dai, H. Improved functional recovery of rat transected spinal cord by peptide-grafted PNIPAM based hydrogel. Colloids Surf. B Biointerfaces 2022, 210, 112220. [Google Scholar] [CrossRef]
  59. Silva, N.A.; Salgado, A.J.; Sousa, R.A.; Oliveira, J.T.; Pedro, A.J.; Leite-Almeida, H.; Cerqueira, R.; Almeida, A.; Mastronardi, F.; Mano, J.F.; et al. Development and Characterization of a Novel Hybrid Tissue Engineering–Based Scaffold for Spinal Cord Injury Repair. Tissue Eng. Part A 2010, 16, 45–54. [Google Scholar] [CrossRef] [Green Version]
  60. Yang, Y.; Fan, Y.; Zhang, H.; Zhang, Q.; Zhao, Y.; Xiao, Z.; Liu, W.; Chen, B.; Gao, L.; Sun, Z.; et al. Small molecules combined with collagen hydrogel direct neurogenesis and migration of neural stem cells after spinal cord injury. Biomaterials 2021, 269, 120479. [Google Scholar] [CrossRef]
  61. Suzuki, H.; Kanchiku, T.; Imajo, Y.; Yoshida, Y.; Nishida, N.; Gondo, T.; Yoshii, S.; Taguchi, T. Artificial collagen-filament scaffold promotes axon regeneration and long tract reconstruction in a rat model of spinal cord transection. Med. Mol. Morphol. 2015, 48, 214–224. [Google Scholar] [CrossRef]
  62. Yara, T.; Kato, Y.; Kataoka, H.; Kanchiku, T.; Suzuki, H.; Gondo, T.; Yoshii, S.; Taguchi, T. Environmental factors involved in axonal regeneration following spinal cord transection in rats. Med. Mol. Morphol. 2009, 42, 150–154. [Google Scholar] [CrossRef] [PubMed]
  63. Nair, L.S.; Laurencin, C.T. Biodegradable polymers as biomaterials. Prog. Polym. Sci. 2007, 32, 762–798. [Google Scholar] [CrossRef]
  64. Cheng, Y.; Zhang, Y.; Wu, H. Polymeric Fibers as Scaffolds for Spinal Cord Injury: A Systematic Review. Front. Bioeng. Biotechnol. 2022, 9, 807533. [Google Scholar] [CrossRef] [PubMed]
  65. Iwasaki, M.; Wilcox, J.T.; Nishimura, Y.; Zweckberger, K.; Suzuki, H.; Wang, J.; Liu, Y.; Karadimas, S.K.; Fehlings, M.G. Synergistic effects of self-assembling peptide and neural stem/progenitor cells to promote tissue repair and forelimb functional recovery in cervical spinal cord injury. Biomaterials 2014, 35, 2617–2629. [Google Scholar] [CrossRef]
  66. Kubinová, Š.; Horák, D.; Hejčl, A.; Plichta, Z.; Kotek, J.; Proks, V.; Forostyak, S.; Syková, E. SIKVAV-modified highly superporous PHEMA scaffolds with oriented pores for spinal cord injury repair. J. Tissue Eng. Regen. Med. 2015, 9, 1298–1309. [Google Scholar] [CrossRef]
  67. Hejčl, A.; Urdzikova, L.M.; Sedy, J.; Lesny, P.; Pradny, M.; Michálek, J.; Burian, M.; Hajek, M.; Zamecnik, J.; Jendelova, P.; et al. Acute and delayed implantation of positively charged 2-hydroxyethyl methacrylate scaffolds in spinal cord injury in the rat. J. Neurosurg. Spine 2008, 8, 67–73. [Google Scholar] [CrossRef] [PubMed]
  68. Sun, F.; Shi, T.; Zhou, T.; Dong, D.; Xie, J.; Wang, R.; An, X.; Chen, M.; Cai, J. 3D Poly(Lactic-co-glycolic acid) Scaffolds for Treating Spinal Cord Injury. J. Biomed. Nanotechnol. 2017, 13, 290–302. [Google Scholar] [CrossRef]
  69. Slotkin, J.R.; Pritchard, C.D.; Luque, B.; Ye, J.; Layer, R.T.; Lawrence, M.S.; O’Shea, T.M.; Roy, R.R.; Zhong, H.; Vollenweider, I.; et al. Biodegradable scaffolds promote tissue remodeling and functional improvement in non-human primates with acute spinal cord injury. Biomaterials 2017, 123, 63–76. [Google Scholar] [CrossRef]
  70. Silva, N.A.; Sousa, R.A.; Fraga, J.S.; Fontes, M.; Leite-Almeida, H.; Cerqueira, R.; Almeida, A.; Sousa, N.; Reis, R.L.; Salgado, A.J. Benefits of Spine Stabilization with Biodegradable Scaffolds in Spinal Cord Injured Rats. Tissue Eng. Part C Methods 2013, 19, 101–108. [Google Scholar] [CrossRef] [Green Version]
  71. Thomas, A.M.; Kubilius, M.B.; Holland, S.J.; Seidlits, S.K.; Boehler, R.M.; Anderson, A.J.; Cummings, B.J.; Shea, L.D. Channel density and porosity of degradable bridging scaffolds on axon growth after spinal injury. Biomaterials 2013, 34, 2213–2220. [Google Scholar] [CrossRef] [Green Version]
  72. Man, W.; Yang, S.; Cao, Z.; Lu, J.; Kong, X.; Sun, X.; Zhao, L.; Guo, Y.; Yao, S.; Wang, G.; et al. A multi-modal delivery strategy for spinal cord regeneration using a composite hydrogel presenting biophysical and biochemical cues synergistically. Biomaterials 2021, 276, 120971. [Google Scholar] [CrossRef] [PubMed]
  73. Kubinová, Š.; Horák, D.; Hejčl, A.; Plichta, Z.; Kotek, J.; Syková, E. Highly superporous cholesterol-modified poly(2-hydroxyethyl methacrylate) scaffolds for spinal cord injury repair. J. Biomed. Mater. Res. Part A 2011, 99A, 618–629. [Google Scholar] [CrossRef] [PubMed]
  74. Guest, J.D.; Moore, S.W.; Aimetti, A.A.; Kutikov, A.B.; Santamaria, A.J.; Hofstetter, C.P.; Ropper, A.E.; Theodore, N.; Ulich, T.R.; Layer, R.T. Internal decompression of the acutely contused spinal cord: Differential effects of irrigation only versus biodegradable scaffold implantation. Biomaterials 2018, 185, 284–300. [Google Scholar] [CrossRef] [PubMed]
  75. Hakim, J.S.; Rodysill, B.R.; Chen, B.K.; Schmeichel, A.M.; Yaszemski, M.J.; Windebank, A.J.; Madigan, N.N. Combinatorial tissue engineering partially restores function after spinal cord injury. J. Tissue Eng. Regen. Med. 2019, 13, 857–873. [Google Scholar] [CrossRef]
  76. Anzalone, A.; Chacko, J.V.; Nishi, R.A.; Dumont, C.; Smith, D.; Shea, L.D.; Digman, M.A.; Cummings, B.J.; Anderson, A.J. Feasibility study on mouse live imaging after spinal cord injury and poly(lactide-co-glycolide) bridge implantation. J. Biomed. Opt. 2018, 23, 065007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. De Laporte, L.; Yan, A.L.; Shea, L.D. Local gene delivery from ECM-coated poly(lactide-co-glycolide) multiple channel bridges after spinal cord injury. Biomaterials 2009, 30, 2361–2368. [Google Scholar] [CrossRef] [Green Version]
  78. Wong, D.Y.; Leveque, J.-C.; Brumblay, H.; Krebsbach, P.H.; Hollister, S.J.; LaMarca, F. Macro-Architectures in Spinal Cord Scaffold Implants Influence Regeneration. J. Neurotrauma 2008, 25, 1027–1037. [Google Scholar] [CrossRef] [Green Version]
  79. Ribeiro-Samy, S.; Silva, N.A.; Correlo, V.M.; Fraga, J.S.; Pinto, L.; Teixeira-Castro, A.; Leite-Almeida, H.; Almeida, A.; Gimble, J.M.; Sousa, N.; et al. Development and Characterization of a PHB-HV-based 3D Scaffold for a Tissue Engineering and Cell-therapy Combinatorial Approach for Spinal Cord Injury Regeneration. Macromol. Biosci. 2013, 13, 1576–1592. [Google Scholar] [CrossRef] [Green Version]
  80. Pawar, K.; Cummings, B.J.; Thomas, A.; Shea, L.D.; Levine, A.; Pfaff, S.; Anderson, A.J. Biomaterial bridges enable regeneration and re-entry of corticospinal tract axons into the caudal spinal cord after SCI: Association with recovery of forelimb function. Biomaterials 2015, 65, 1–12. [Google Scholar] [CrossRef] [Green Version]
  81. Rooney, G.E.; Vaishya, S.; Ameenuddin, S.; Currier, B.L.; Schiefer, T.K.; Knight, A.; Chen, B.; Mishra, P.K.; Spinner, R.J.; Macura, S.I.; et al. Rigid Fixation of the Spinal Column Improves Scaffold Alignment and Prevents Scoliosis in the Transected Rat Spinal Cord. Spine 2008, 33, E914–E919. [Google Scholar] [CrossRef] [PubMed]
  82. Shu, B.; Sun, X.; Liu, R.; Jiang, F.; Yu, H.; Xu, N.; An, Y. Restoring electrical connection using a conductive biomaterial provides a new therapeutic strategy for rats with spinal cord injury. Neurosci. Lett. 2019, 692, 33–40. [Google Scholar] [CrossRef] [PubMed]
  83. Zhou, L.; Fan, L.; Yi, X.; Zhou, Z.; Liu, C.; Fu, R.; Dai, C.; Wang, Z.; Chen, X.; Yu, P.; et al. Soft Conducting Polymer Hydrogels Cross-Linked and Doped by Tannic Acid for Spinal Cord Injury Repair. ACS Nano 2018, 12, 10957–10967. [Google Scholar] [CrossRef] [PubMed]
  84. Pertici, V.; Trimaille, T.; Laurin, J.; Felix, M.-S.; Marqueste, T.; Pettmann, B.; Chauvin, J.-P.; Gigmes, D.; Decherchi, P. Repair of the injured spinal cord by implantation of a synthetic degradable block copolymer in rat. Biomaterials 2014, 35, 6248–6258. [Google Scholar] [CrossRef] [PubMed]
  85. Reis, K.P.; Sperling, L.E.; Teixeira, C.; Sommer, L.; Colombo, M.; Koester, L.S.; Pranke, P. VPA/PLGA microfibers produced by coaxial electrospinning for the treatment of central nervous system injury. Braz. J. Med. Biol. Res. 2020, 53, e8993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Novikova, L.N.; Kolar, M.K.; Kingham, P.J.; Ullrich, A.; Oberhoffner, S.; Renardy, M.; Doser, M.; Müller, E.; Wiberg, M.; Novikov, L.N. Trimethylene carbonate-caprolactone conduit with poly-p-dioxanone microfilaments to promote regeneration after spinal cord injury. Acta Biomater. 2018, 66, 177–191. [Google Scholar] [CrossRef] [PubMed]
  87. Song, Y.H.; Agrawal, N.K.; Griffin, J.M.; Schmidt, C.E. Recent advances in nanotherapeutic strategies for spinal cord injury repair. Adv. Drug Deliv. Rev. 2019, 148, 38–59. [Google Scholar] [CrossRef] [PubMed]
  88. Gerardo-Nava, J.; Führmann, T.; Klinkhammer, K.; Seiler, N.; Mey, J.; Klee, D.; Möller, M.; Dalton, P.D.; Brook, G.A. Human neural cell interactions with orientated electrospun nanofibers in vitro. Nanomedicine 2009, 4, 11–30. [Google Scholar] [CrossRef]
  89. Zamani, F.; Amani-Tehran, M.; Latifi, M.; Shokrgozar, M.A.; Zaminy, A. Promotion of spinal cord axon regeneration by 3D nanofibrous core-sheath scaffolds. J. Biomed. Mater. Res. Part A 2014, 102, 506–513. [Google Scholar] [CrossRef]
  90. Sun, X.; Bai, Y.; Zhai, H.; Liu, S.; Zhang, C.; Xu, Y.; Zou, J.; Wang, T.; Chen, S.; Zhu, Q.; et al. Devising micro/nano-architectures in multi-channel nerve conduits towards a pro-regenerative matrix for the repair of spinal cord injury. Acta Biomater. 2019, 86, 194–206. [Google Scholar] [CrossRef]
  91. Cigognini, D.; Silva, D.; Paloppi, S.; Gelain, F. Evaluation of mechanical properties and therapeutic effect of injectable self-assembling hydrogels for spinal cord injury. J. Biomed. Nanotechnol. 2014, 10, 309–323. [Google Scholar] [CrossRef]
  92. Yao, S.; Yu, S.; Cao, Z.; Yang, Y.; Yu, X.; Mao, H.-Q.; Wang, L.-N.; Sun, X.; Zhao, L.; Wang, X. Hierarchically aligned fibrin nanofiber hydrogel accelerated axonal regrowth and locomotor function recovery in rat spinal cord injury. Int. J. Nanomed. 2018, 13, 2883–2895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Altinova, H.; Möllers, S.; Deumens, R.; Gerardo-Nava, J.; Führmann, T.; van Neerven, S.G.A.; Bozkurt, A.; Mueller, C.A.; Hoff, H.J.; Heschel, I.; et al. Functional recovery not correlated with axon regeneration through olfactory ensheathing cell-seeded scaffolds in a model of acute spinal cord injury. Tissue Eng. Regen. Med. 2016, 13, 585–600. [Google Scholar] [CrossRef]
  94. Usmani, S.; Biagioni, A.F.; Medelin, M.; Scaini, D.; Casani, R.; Aurand, E.R.; Padro, D.; Egimendia, A.; Cabrer, P.R.; Scarselli, M.; et al. Functional rewiring across spinal injuries via biomimetic nanofiber scaffolds. Proc. Natl. Acad. Sci. USA 2020, 117, 25212–25218. [Google Scholar] [CrossRef]
  95. Sever-Bahcekapili, M.; Yilmaz, C.; Demirel, A.; Kilinc, M.C.; Dogan, I.; Caglar, Y.S.; Guler, M.O.; Tekinay, A.B. Neuroactive Peptide Nanofibers for Regeneration of Spinal Cord after Injury. Macromol. Biosci. 2021, 21, e2000234. [Google Scholar] [CrossRef] [PubMed]
  96. Zhao, T.; Jing, Y.; Zhou, X.; Wang, J.; Huang, X.; Gao, L.; Zhu, Y.; Wang, L.; Gou, Z.; Liang, C.; et al. PHBV/PLA/Col-Based Nanofibrous Scaffolds Promote Recovery of Locomotor Function by Decreasing Reactive Astrogliosis in a Hemisection Spinal Cord Injury Rat Model. J. Biomed. Nanotechnol. 2018, 14, 1921–1933. [Google Scholar] [CrossRef] [PubMed]
  97. Chedly, J.; Soares, S.; Montembault, A.; von Boxberg, Y.; Veron-Ravaille, M.; Mouffle, C.; Benassy, M.-N.; Taxi, J.; David, L.; Nothias, F. Physical chitosan microhydrogels as scaffolds for spinal cord injury restoration and axon regeneration. Biomaterials 2017, 138, 91–107. [Google Scholar] [CrossRef] [Green Version]
  98. Cigognini, D.; Satta, A.; Colleoni, B.; Silva, D.; Donegà, M.; Antonini, S.; Gelain, F. Evaluation of Early and Late Effects into the Acute Spinal Cord Injury of an Injectable Functionalized Self-Assembling Scaffold. PLoS ONE 2011, 6, e19782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Palejwala, A.H.; Fridley, J.S.; Mata, J.A.; Samuel, E.L.G.; Luerssen, T.G.; Perlaky, L.; Kent, T.A.; Tour, J.M.; Jea, A. Biocompatibility of reduced graphene oxide nanoscaffolds following acute spinal cord injury in rats. Surg. Neurol. Int. 2016, 7, 75. [Google Scholar] [CrossRef] [Green Version]
  100. Pawelec, K.M.; Koffler, J.; Shahriari, D.; Galvan, A.; Tuszynski, M.H.; Sakamoto, J. Microstructure and in vivo characterization of multi-channel nerve guidance scaffolds. Biomed. Mater. 2018, 13, 044104. [Google Scholar] [CrossRef]
  101. Milbreta, U.; Nguyen, L.H.; Diao, H.; Lin, J.; Wu, W.; Sun, C.-Y.; Wang, J.; Chew, S.Y. Three-Dimensional Nanofiber Hybrid Scaffold Directs and Enhances Axonal Regeneration after Spinal Cord Injury. ACS Biomater. Sci. Eng. 2016, 2, 1319–1329. [Google Scholar] [CrossRef]
  102. Tysseling, V.M.; Sahni, V.; Pashuck, E.T.; Birch, D.; Hebert, A.; Czeisler, C.; Stupp, S.I.; Kessler, J.A. Self-assembling peptide amphiphile promotes plasticity of serotonergic fibers following spinal cord injury. J. Neurosci. Res. 2010, 88, 3161–3170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Liu, Y.; Ye, H.; Satkunendrarajah, K.; Yao, G.S.; Bayon, Y.; Fehlings, M.G. A self-assembling peptide reduces glial scarring, attenuates post-traumatic inflammation and promotes neurological recovery following spinal cord injury. Acta Biomater. 2013, 9, 8075–8088. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, Y.; Tan, H.; Hui, X. Biomaterial Scaffolds in Regenerative Therapy of the Central Nervous System. BioMed Res. Int. 2018, 2018, 7848901. [Google Scholar] [CrossRef] [Green Version]
  105. Furuya, T.; Hashimoto, M.; Koda, M.; Murata, A.; Okawa, A.; Dezawa, M.; Matsuse, D.; Tabata, Y.; Takahashi, K.; Yamazaki, M. Treatment with basic fibroblast growth factor-incorporated gelatin hydrogel does not exacerbate mechanical allodynia after spinal cord contusion injury in rats. J. Spinal Cord Med. 2013, 36, 134–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chvatal, S.A.; Kim, Y.-T.; Bratt-Leal, A.M.; Lee, H.; Bellamkonda, R.V. Spatial distribution and acute anti-inflammatory effects of Methylprednisolone after sustained local delivery to the contused spinal cord. Biomaterials 2008, 29, 1967–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. McKay, C.A.; Pomrenke, R.D.; McLane, J.S.; Schaub, N.J.; DeSimone, E.K.; Ligon, L.A.; Gilbert, R.J. An Injectable, Calcium Responsive Composite Hydrogel for the Treatment of Acute Spinal Cord Injury. ACS Appl. Mater. Interfaces 2014, 6, 1424–1438. [Google Scholar] [CrossRef] [PubMed]
  108. Jain, A.; McKeon, R.J.; Brady-Kalnay, S.M.; Bellamkonda, R.V. Sustained Delivery of Activated Rho GTPases and BDNF Promotes Axon Growth in CSPG-Rich Regions Following Spinal Cord Injury. PLoS ONE 2011, 6, e16135. [Google Scholar] [CrossRef] [Green Version]
  109. Wen, Y.; Yu, S.; Wu, Y.; Ju, R.; Wang, H.; Liu, Y.; Wang, Y.; Xu, Q. Spinal cord injury repair by implantation of structured hyaluronic acid scaffold with PLGA microspheres in the rat. Cell Tissue Res. 2016, 364, 17–28. [Google Scholar] [CrossRef]
  110. Chen, B.; He, J.; Yang, H.; Zhang, Q.; Zhang, L.; Zhang, X.; Xie, E.; Liu, C.; Zhang, R.; Wang, Y.; et al. Repair of spinal cord injury by implantation of bFGF-incorporated HEMA-MOETACL hydrogel in rats. Sci. Rep. 2015, 5, 9017. [Google Scholar] [CrossRef] [Green Version]
  111. Chan, G.; Mooney, D.J. New materials for tissue engineering: Towards greater control over the biological response. Trends Biotechnol. 2008, 26, 382–392. [Google Scholar] [CrossRef]
  112. Straley, K.S.; Foo, C.W.P.; Heilshorn, S.C. Biomaterial Design Strategies for the Treatment of Spinal Cord Injuries. J. Neurotrauma 2010, 27, 1–19. [Google Scholar] [CrossRef]
  113. Kalotra, S.; Saini, V.; Singh, H.; Sharma, A.; Kaur, G. 5-Nonyloxytryptamine oxalate–embedded collagen–laminin scaffolds augment functional recovery after spinal cord injury in mice. Ann. N. Y. Acad. Sci. 2020, 1465, 99–116. [Google Scholar] [CrossRef] [PubMed]
  114. Lin, J.; Anopas, D.; Milbreta, U.; Lin, P.H.; Chin, J.S.; Zhang, N.; Wee, S.K.; Tow, A.; Ang, W.T.; Chew, S.Y. Regenerative rehabilitation: Exploring the synergistic effects of rehabilitation and implantation of a bio-functional scaffold in enhancing nerve regeneration. Biomater. Sci. 2019, 7, 5150–5160. [Google Scholar] [CrossRef] [PubMed]
  115. Shi, Q.; Gao, W.; Han, X.; Zhu, X.; Sun, J.; Xie, F.; Hou, X.; Yang, H.; Dai, J.; Chen, L. Collagen scaffolds modified with collagen-binding bFGF promotes the neural regeneration in a rat hemisected spinal cord injury model. Sci. China Life Sci. 2014, 57, 232–240. [Google Scholar] [CrossRef] [Green Version]
  116. Wang, X.; Li, Y.; Gao, Y.; Chen, X.; Yao, J.; Lin, W.; Chen, Y.; Liu, J.; Yang, Y.; Wang, X. Combined use of spinal cord-mimicking partition type scaffold architecture and neurotrophin-3 for surgical repair of completely transected spinal cord in rats. J. Biomater. Sci. Polym. Ed. 2013, 24, 927–939. [Google Scholar] [CrossRef] [PubMed]
  117. Li, G.; Che, M.-T.; Zhang, K.; Qin, L.-N.; Zhang, Y.-T.; Chen, R.-Q.; Rong, L.-M.; Liu, S.; Ding, Y.; Shen, H.-Y.; et al. Graft of the NT-3 persistent delivery gelatin sponge scaffold promotes axon regeneration, attenuates inflammation, and induces cell migration in rat and canine with spinal cord injury. Biomaterials 2016, 83, 233–248. [Google Scholar] [CrossRef] [PubMed]
  118. Wei, Y.-T.; He, Y.; Xu, C.-L.; Wang, Y.; Liu, B.-F.; Wang, X.-M.; Sun, X.-D.; Cui, F.-Z.; Xu, Q.-Y. Hyaluronic acid hydrogel modified with nogo-66 receptor antibody and poly-L-lysine to promote axon regrowth after spinal cord injury. J. Biomed. Mater. Res. Part B Appl. Biomater. 2010, 95B, 110–117. [Google Scholar] [CrossRef] [PubMed]
  119. Bighinati, A.; Focarete, M.L.; Gualandi, C.; Pannella, M.; Giuliani, A.; Beggiato, S.; Ferraro, L.; Lorenzini, L.; Giardino, L.; Calzà, L. Improved Functional Recovery in Rat Spinal Cord Injury Induced by a Drug Combination Administered with an Implantable Polymeric Delivery System. J. Neurotrauma 2020, 37, 1708–1719. [Google Scholar] [CrossRef]
  120. Ehsanipour, A.; Sathialingam, M.; Rad, L.M.; de Rutte, J.; Bierman, R.D.; Liang, J.; Xiao, W.; Di Carlo, D.; Seidlits, S.K. Injectable, macroporous scaffolds for delivery of therapeutic genes to the injured spinal cord. APL Bioeng. 2021, 5, 016104. [Google Scholar] [CrossRef]
  121. Xie, J.; Li, J.; Ma, J.; Li, M.; Wang, X.; Fu, X.; Ma, Y.; Yang, H.; Li, B. Saijilafu Magnesium Oxide/Poly(l-lactide-co-ε-caprolactone) Scaffolds Loaded with Neural Morphogens Promote Spinal Cord Repair through Targeting the Calcium Influx and Neuronal Differentiation of Neural Stem Cells. Adv. Healthc. Mater. 2022, 11, 2200386. [Google Scholar] [CrossRef]
  122. Xi, K.; Gu, Y.; Tang, J.; Chen, H.; Xu, Y.; Wu, L.; Cai, F.; Deng, L.; Yang, H.; Shi, Q.; et al. Microenvironment-responsive immunoregulatory electrospun fibers for promoting nerve function recovery. Nat. Commun. 2020, 11, 4504, Erratum in Nat. Commun. 2021, 12, 2882. [Google Scholar] [CrossRef]
  123. Rooney, G.E.; Knight, A.M.; Madigan, N.N.; Gross, L.; Chen, B.; Giraldo, C.V.; Seo, S.; Nesbitt, J.J.; Dadsetan, M.; Yaszemski, M.J.; et al. Sustained Delivery of Dibutyryl Cyclic Adenosine Monophosphate to the Transected Spinal Cord Via Oligo [(Polyethylene Glycol) Fumarate] Hydrogels. Tissue Eng. Part A 2011, 17, 1287–1302. [Google Scholar] [CrossRef]
  124. Stropkovska, A.; Kisucka, A.; Bimbova, K.; Bacova, M.; Galik, J.; Medvecky, L.; Sulla, I.; Karasova, M.; Lukacova, N. Combined therapy (Rho-A-kinase inhibitor and chitosan/collagen porous scaffold) provides a supportive environment for endogenous regenerative processes after spinal cord trauma. Arch. Ital. Biol. 2021, 159, 159–177. [Google Scholar] [CrossRef]
  125. Ji, W.-C.; Li, M.; Jiang, W.-T.; Ma, X.; Li, J. Protective effect of brain-derived neurotrophic factor and neurotrophin-3 overexpression by adipose-derived stem cells combined with silk fibroin/chitosan scaffold in spinal cord injury. Neurol. Res. 2020, 42, 361–371. [Google Scholar] [CrossRef]
  126. Katz, J.S.; Burdick, J.A. Hydrogel mediated delivery of trophic factors for neural repair. WIREs Nanomed. Nanobiotechnol. 2008, 1, 128–139. [Google Scholar] [CrossRef]
  127. Johnson, P.J.; Tatara, A.; McCreedy, D.A.; Shiu, A.; Sakiyama-Elbert, S.E. Tissue-engineered fibrin scaffolds containing neural progenitors enhance functional recovery in a subacute model of SCI. Soft Matter 2010, 6, 5127–5137. [Google Scholar] [CrossRef] [Green Version]
  128. Smith, D.R.; Dumont, C.M.; Park, J.; Ciciriello, A.J.; Guo, A.; Tatineni, R.; Cummings, B.J.; Anderson, A.J.; Shea, L.D. Polycistronic Delivery of IL-10 and NT-3 Promotes Oligodendrocyte Myelination and Functional Recovery in a Mouse Spinal Cord Injury Model. Tissue Eng. Part A 2020, 26, 672–682. [Google Scholar] [CrossRef]
  129. Jain, A.; Kim, Y.-T.; McKeon, R.J.; Bellamkonda, R.V. In situ gelling hydrogels for conformal repair of spinal cord defects, and local delivery of BDNF after spinal cord injury. Biomaterials 2006, 27, 497–504. [Google Scholar] [CrossRef]
  130. Breen, B.A.; Kraskiewicz, H.; Ronan, R.; Kshiragar, A.; Patar, A.; Sargeant, T.; Pandit, A.; McMahon, S.S. Therapeutic Effect of Neurotrophin-3 Treatment in an Injectable Collagen Scaffold Following Rat Spinal Cord Hemisection Injury. ACS Biomater. Sci. Eng. 2017, 3, 1287–1295. [Google Scholar] [CrossRef] [Green Version]
  131. Rosenzweig, E.S.; Brock, J.H.; Lu, P.; Kumamaru, H.; Salegio, E.A.; Kadoya, K.; Weber, J.L.; Liang, J.J.; Moseanko, R.; Hawbecker, S.; et al. Restorative effects of human neural stem cell grafts on the primate spinal cord. Nat. Med. 2018, 24, 484–490. [Google Scholar] [CrossRef]
  132. Zhao, Y.; Tang, F.; Xiao, Z.; Han, G.; Wang, N.; Yin, N.; Chen, B.; Jiang, X.; Yun, C.; Han, W.; et al. Clinical Study of NeuroRegen Scaffold Combined with Human Mesenchymal Stem Cells for the Repair of Chronic Complete Spinal Cord Injury. Cell Transplant. 2017, 26, 891–900. [Google Scholar] [CrossRef]
  133. Chen, W.; Zhang, Y.; Yang, S.; Sun, J.; Qiu, H.; Hu, X.; Niu, X.; Xiao, Z.; Zhao, Y.; Zhou, Y.; et al. NeuroRegen Scaffolds Combined with Autologous Bone Marrow Mononuclear Cells for the Repair of Acute Complete Spinal Cord Injury: A 3-Year Clinical Study. Cell Transplant. 2020, 29, 963689720950637. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, L.; Chueng, S.-T.D.; Li, Y.; Patel, M.; Rathnam, C.; Dey, G.; Wang, L.; Cai, L.; Lee, K.-B. A biodegradable hybrid inorganic nanoscaffold for advanced stem cell therapy. Nat. Commun. 2018, 9, 3147. [Google Scholar] [CrossRef] [Green Version]
  135. Guo, J.; Su, H.; Zeng, Y.; Liang, Y.-X.; Wong, W.M.; Ellis-Behnke, R.G.; So, K.-F.; Wu, W. Reknitting the injured spinal cord by self-assembling peptide nanofiber scaffold. Nanomed. Nanotechnol. Biol. Med. 2007, 3, 311–321. [Google Scholar] [CrossRef]
  136. Kim, S.-H.; Hwang, K.; Lee, H.A.; Kim, J.; Cho, M.; Kim, M.; Shin, J.E.; Lee, H.; Park, K.I.; Jang, J.-H. Pastable, Adhesive, Injectable, Nanofibrous, and Tunable (PAINT) Biphasic Hybrid Matrices as Versatile Therapeutic Carriers. ACS Appl. Mater. Interfaces 2021, 13, 42429–42441. [Google Scholar] [CrossRef]
  137. Xu, B.; Zhao, Y.; Xiao, Z.; Wang, B.; Liang, H.; Li, X.; Fang, Y.; Han, S.; Li, X.; Fan, C.; et al. A Dual Functional Scaffold Tethered with EGFR Antibody Promotes Neural Stem Cell Retention and Neuronal Differentiation for Spinal Cord Injury Repair. Adv. Healthc. Mater. 2017, 6, 1601279. [Google Scholar] [CrossRef]
  138. Li, X.; Xiao, Z.; Han, J.; Chen, L.; Xiao, H.; Ma, F.; Hou, X.; Li, X.; Sun, J.; Ding, W.; et al. Promotion of neuronal differentiation of neural progenitor cells by using EGFR antibody functionalized collagen scaffolds for spinal cord injury repair. Biomaterials 2013, 34, 5107–5116. [Google Scholar] [CrossRef]
  139. Tang, S.; Liao, X.; Shi, B.; Qu, Y.; Huang, Z.; Lin, Q.; Guo, X.; Pei, F. The Effects of Controlled Release of Neurotrophin-3 from PCLA Scaffolds on the Survival and Neuronal Differentiation of Transplanted Neural Stem Cells in a Rat Spinal Cord Injury Model. PLoS ONE 2014, 9, e107517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Yu, D.; Neeley, W.L.; Pritchard, C.D.; Slotkin, J.R.; Woodard, E.J.; Langer, R.; Teng, Y.D. Blockade of Peroxynitrite-Induced Neural Stem Cell Death in the Acutely Injured Spinal Cord by Drug-Releasing Polymer. Stem Cells 2009, 27, 1212–1222, Erratum in Stem Cells 2009, 27, 3121. [Google Scholar] [CrossRef] [Green Version]
  141. Ham, T.R.; Pukale, D.D.; Hamrangsekachaee, M.; Leipzig, N.D. Subcutaneous priming of protein-functionalized chitosan scaffolds improves function following spinal cord injury. Mater. Sci. Eng. C 2020, 110, 110656. [Google Scholar] [CrossRef]
  142. Liu, X.; Hao, M.; Chen, Z.; Zhang, T.; Huang, J.; Dai, J.; Zhang, Z. 3D bioprinted neural tissue constructs for spinal cord injury repair. Biomaterials 2021, 272, 120771. [Google Scholar] [CrossRef] [PubMed]
  143. Rauch, M.F.; Hynes, S.R.; Bertram, J.; Redmond, A.; Robinson, R.; Williams, C.; Xu, H.; Madri, J.A.; Lavik, E.B. Engineering angiogenesis following spinal cord injury: A coculture of neural progenitor and endothelial cells in a degradable polymer implant leads to an increase in vessel density and formation of the blood-spinal cord barrier. Eur. J. Neurosci. 2009, 29, 132–145. [Google Scholar] [CrossRef] [PubMed]
  144. Li, X.; Fan, C.; Xiao, Z.; Zhao, Y.; Zhang, H.; Sun, J.; Zhuang, Y.; Wu, X.; Shi, J.; Chen, Y.; et al. A collagen microchannel scaffold carrying paclitaxel-liposomes induces neuronal differentiation of neural stem cells through Wnt/β-catenin signaling for spinal cord injury repair. Biomaterials 2018, 183, 114–127. [Google Scholar] [CrossRef] [PubMed]
  145. Liu, S.; Xie, Y.-Y.; Wang, L.-D.; Tai, C.-X.; Chen, D.; Mu, D.; Cui, Y.-Y.; Wang, B. A multi-channel collagen scaffold loaded with neural stem cells for the repair of spinal cord injury. Neural Regen. Res. 2021, 16, 2284–2292. [Google Scholar] [CrossRef] [PubMed]
  146. Zou, Y.; Ma, D.; Shen, H.; Zhao, Y.; Xu, B.; Fan, Y.; Sun, Z.; Chen, B.; Xue, W.; Shi, Y.; et al. Aligned collagen scaffold combination with human spinal cord-derived neural stem cells to improve spinal cord injury repair. Biomater. Sci. 2020, 8, 5145–5156. [Google Scholar] [CrossRef]
  147. Martínez-Ramos, C.; Doblado, L.R.; Mocholi, E.L.; Alastrue-Agudo, A.; Petidier, M.S.; Giraldo, E.; Pradas, M.M.; Moreno-Manzano, V. Biohybrids for spinal cord injury repair. J. Tissue Eng. Regen. Med. 2019, 13, 509–521. [Google Scholar] [CrossRef]
  148. Shin, J.E.; Jung, K.; Kim, M.; Hwang, K.; Lee, H.; Kim, I.-S.; Lee, B.H.; Lee, I.-S.; Park, K.I. Brain and spinal cord injury repair by implantation of human neural progenitor cells seeded onto polymer scaffolds. Exp. Mol. Med. 2018, 50, 1–18. [Google Scholar] [CrossRef] [Green Version]
  149. Nomura, H.; Zahir, T.; Kim, H.; Katayama, Y.; Kulbatski, I.; Morshead, C.M.; Shoichet, M.S.; Tator, C.H. Extramedullary Chitosan Channels Promote Survival of Transplanted Neural Stem and Progenitor Cells and Create a Tissue Bridge After Complete Spinal Cord Transection. Tissue Eng. Part A 2008, 14, 649–665. [Google Scholar] [CrossRef] [Green Version]
  150. Ma, Y.-H.; Shi, H.-J.; Wei, Q.-S.; Deng, Q.-W.; Sun, J.-H.; Liu, Z.; Lai, B.-Q.; Li, G.; Ding, Y.; Niu, W.-T.; et al. Developing a mechanically matched decellularized spinal cord scaffold for the in situ matrix-based neural repair of spinal cord injury. Biomaterials 2021, 279, 121192. [Google Scholar] [CrossRef]
  151. Chen, G.; Hu, Y.R.; Wan, H.; Deng, Q.W.; Sun, J.H.; Liu, Z.; Lai, B.Q.; Li, G.; Ding, Y.; Niu, W.T.; et al. Functional recovery following traumatic spinal cord injury mediated by a unique polymer scaffold seeded with neural stem cells and Schwann cells. Chin. Med. J. 2010, 123, 2424–2431. [Google Scholar]
  152. Ruzicka, J.; Romanyuk, N.; Jirakova, K.; Hejcl, A.; Janouskova, O.; Machova, L.U.; Bochin, M.; Pradny, M.; Vargova, L.; Jendelova, P. The Effect of iPS-Derived Neural Progenitors Seeded on Laminin-Coated pHEMA-MOETACl Hydrogel with Dual Porosity in a Rat Model of Chronic Spinal Cord Injury. Cell Transplant. 2019, 28, 400–412. [Google Scholar] [CrossRef] [Green Version]
  153. Hu, Y.; Zhang, F.; Zhong, W.; Liu, Y.; He, Q.; Yang, M.; Chen, H.; Xu, X.; Bian, K.; Xu, J.; et al. Transplantation of neural scaffolds consisting of dermal fibroblast-reprogrammed neurons and 3D silk fibrous materials promotes the repair of spinal cord injury. J. Mater. Chem. B 2019, 7, 7525–7539. [Google Scholar] [CrossRef]
  154. Olson, H.E.; Rooney, G.E.; Gross, L.; Nesbitt, J.J.; Galvin, K.E.; Knight, A.; Chen, B.; Yaszemski, M.J.; Windebank, A.J. Neural Stem Cell– and Schwann Cell–Loaded Biodegradable Polymer Scaffolds Support Axonal Regeneration in the Transected Spinal Cord. Tissue Eng. Part A 2009, 15, 1797–1805. [Google Scholar] [CrossRef]
  155. Kourgiantaki, A.; Tzeranis, D.S.; Karali, K.; Georgelou, K.; Bampoula, E.; Psilodimitrakopoulos, S.; Yannas, I.V.; Stratakis, E.; Sidiropoulou, K.; Charalampopoulos, I.; et al. Neural stem cell delivery via porous collagen scaffolds promotes neuronal differentiation and locomotion recovery in spinal cord injury. NPJ Regen. Med. 2020, 5, 12. [Google Scholar] [CrossRef] [PubMed]
  156. Xia, L.; Wan, H.; Hao, S.-Y.; Li, D.-Z.; Chen, G.; Gao, C.-C.; Li, J.-H.; Yang, F.; Wang, S.-G.; Liu, S. Co-transplantation of neural stem cells and Schwann cells within poly (L-lactic-co-glycolic acid) scaffolds facilitates axonal regeneration in hemisected rat spinal cord. Chin. Med. J. 2013, 126, 909–917. [Google Scholar] [PubMed]
  157. Zarei-Kheirabadi, M.; Sadrosadat, H.; Mohammadshirazi, A.; Jaberi, R.; Sorouri, F.; Khayyatan, F.; Kiani, S. Human embryonic stem cell-derived neural stem cells encapsulated in hyaluronic acid promotes regeneration in a contusion spinal cord injured rat. Int. J. Biol. Macromol. 2020, 148, 1118–1129. [Google Scholar] [CrossRef]
  158. Sugai, K.; Nishimura, S.; Kato-Negishi, M.; Onoe, H.; Iwanaga, S.; Toyama, Y.; Matsumoto, M.; Takeuchi, S.; Okano, H.; Nakamura, M. Neural stem/progenitor cell-laden microfibers promote transplant survival in a mouse transected spinal cord injury model. J. Neurosci. Res. 2015, 93, 1826–1838. [Google Scholar] [CrossRef]
  159. Kim, B.G.; Kang, Y.M.; Phi, J.H.; Kim, Y.-H.; Hwang, D.H.; Choi, J.Y.; Ryu, S.; Elastal, A.-E.; Paek, S.H.; Wang, K.-C.; et al. Implantation of polymer scaffolds seeded with neural stem cells in a canine spinal cord injury model. Cytotherapy 2010, 12, 841–845. [Google Scholar] [CrossRef]
  160. McCreedy, D.A.; Wilems, T.S.; Xu, H.; Butts, J.C.; Brown, C.R.; Smith, A.W.; Sakiyama-Elbert, S.E. Survival, differentiation, and migration of high-purity mouse embryonic stem cell-derived progenitor motor neurons in fibrin scaffolds after sub-acute spinal cord injury. Biomater. Sci. 2014, 2, 1672–1682. [Google Scholar] [CrossRef] [Green Version]
  161. Jian, R.; Yixu, Y.; Sheyu, L.; Jianhong, S.; Yaohua, Y.; Xing, S.; Qingfeng, H.; Xiaojian, L.; Lei, Z.; Yan, Z.; et al. Repair of spinal cord injury by chitosan scaffold with glioma ECM and SB216763 implantation in adult rats. J. Biomed. Mater. Res. Part A 2015, 103, 3259–3272. [Google Scholar] [CrossRef] [PubMed]
  162. Wang, J.; Chu, R.; Ni, N.; Nan, G. The effect of Matrigel as scaffold material for neural stem cell transplantation for treating spinal cord injury. Sci. Rep. 2020, 10, 2576. [Google Scholar] [CrossRef] [Green Version]
  163. Zou, Y.; Zhao, Y.; Xiao, Z.; Chen, B.; Ma, D.; Shen, H.; Gu, R.; Dai, J. Comparison of Regenerative Effects of Transplanting Three-Dimensional Longitudinal Scaffold Loaded-Human Mesenchymal Stem Cells and Human Neural Stem Cells on Spinal Cord Completely Transected Rats. ACS Biomater. Sci. Eng. 2020, 6, 1671–1680. [Google Scholar] [CrossRef]
  164. Yang, L.; Conley, B.M.; Cerqueira, S.R.; Pongkulapa, T.; Wang, S.; Lee, J.K.; Lee, K.-B. Effective Modulation of CNS Inhibitory Microenvironment using Bioinspired Hybrid-Nanoscaffold-Based Therapeutic Interventions. Adv. Mater. 2020, 32, 2002578. [Google Scholar] [CrossRef]
  165. Nori, S.; Khazaei, M.; Ahuja, C.S.; Yokota, K.; Ahlfors, J.-E.; Liu, Y.; Wang, J.; Shibata, S.; Chio, J.; Hettiaratchi, M.H.; et al. Human Oligodendrogenic Neural Progenitor Cells Delivered with Chondroitinase ABC Facilitate Functional Repair of Chronic Spinal Cord Injury. Stem Cell Rep. 2018, 11, 1433–1448. [Google Scholar] [CrossRef] [Green Version]
  166. Sun, X.; Zhang, C.; Xu, J.; Zhai, H.; Liu, S.; Xu, Y.; Hu, Y.; Long, H.; Bai, Y.; Quan, D. Neurotrophin-3-Loaded Multichannel Nanofibrous Scaffolds Promoted Anti-Inflammation, Neuronal Differentiation, and Functional Recovery after Spinal Cord Injury. ACS Biomater. Sci. Eng. 2020, 6, 1228–1238. [Google Scholar] [CrossRef]
  167. Hatami, M.; Mehrjardi, N.Z.; Kiani, S.; Hemmesi, K.; Azizi, H.; Shahverdi, A.; Baharvand, H. Human embryonic stem cell-derived neural precursor transplants in collagen scaffolds promote recovery in injured rat spinal cord. Cytotherapy 2009, 11, 618–630. [Google Scholar] [CrossRef]
  168. Lu, P.; Graham, L.; Wang, Y.; Wu, D.; Tuszynski, M. Promotion of Survival and Differentiation of Neural Stem Cells with Fibrin and Growth Factor Cocktails after Severe Spinal Cord Injury. J. Vis. Exp. 2014, e50641. [Google Scholar] [CrossRef] [Green Version]
  169. Lu, P.; Wang, Y.; Graham, L.; McHale, K.; Gao, M.; Wu, D.; Brock, J.; Blesch, A.; Rosenzweig, E.S.; Havton, L.A.; et al. Long-Distance Growth and Connectivity of Neural Stem Cells after Severe Spinal Cord Injury. Cell 2012, 150, 1264–1273. [Google Scholar] [CrossRef] [Green Version]
  170. Mothe, A.J.; Tam, R.Y.; Zahir, T.; Tator, C.H.; Shoichet, M.S. Repair of the injured spinal cord by transplantation of neural stem cells in a hyaluronan-based hydrogel. Biomaterials 2013, 34, 3775–3783. [Google Scholar] [CrossRef]
  171. Führmann, T.; Tam, R.Y.; Ballarin, B.; Coles, B.; Donaghue, I.E.; van der Kooy, D.; Nagy, A.; Tator, C.H.; Morshead, C.M.; Shoichet, M.S. Injectable hydrogel promotes early survival of induced pluripotent stem cell-derived oligodendrocytes and attenuates longterm teratoma formation in a spinal cord injury model. Biomaterials 2016, 83, 23–36. [Google Scholar] [CrossRef]
  172. Koffler, J.; Zhu, W.; Qu, X.; Platoshyn, O.; Dulin, J.N.; Brock, J.; Graham, L.; Lu, P.; Sakamoto, J.; Marsala, M.; et al. Biomimetic 3D-printed scaffolds for spinal cord injury repair. Nat. Med. 2019, 25, 263–269. [Google Scholar] [CrossRef]
  173. Fan, L.; Liu, C.; Chen, X.; Zou, Y.; Zhou, Z.; Lin, C.; Tan, G.; Zhou, L.; Ning, C.; Wang, Q. Directing Induced Pluripotent Stem Cell Derived Neural Stem Cell Fate with a Three-Dimensional Biomimetic Hydrogel for Spinal Cord Injury Repair. ACS Appl. Mater. Interfaces 2018, 10, 17742–17755. [Google Scholar] [CrossRef]
  174. Zhang, L.; Fan, C.; Hao, W.; Zhuang, Y.; Liu, X.; Zhao, Y.; Chen, B.; Xiao, Z.; Chen, Y.; Dai, J. NSCs Migration Promoted and Drug Delivered Exosomes-Collagen Scaffold via a Bio-Specific Peptide for One-Step Spinal Cord Injury Repair. Adv. Healthc. Mater. 2021, 10, e2001896. [Google Scholar] [CrossRef]
  175. Zhou, X.; Shi, G.; Fan, B.; Cheng, X.; Zhang, X.; Wang, X.; Liu, S.; Hao, Y.; Wei, Z.; Wang, L.; et al. Polycaprolactone electrospun fiber scaffold loaded with iPSCs-NSCs and ASCs as a novel tissue engineering scaffold for the treatment of spinal cord injury. Int. J. Nanomed. 2018, 13, 6265–6277. [Google Scholar] [CrossRef] [Green Version]
  176. Shao, R.; Li, C.; Chen, Y.; Zhang, L.; Yang, H.; Zhang, Z.; Yue, J.; Gao, W.; Zhu, H.; Pan, H.; et al. LncRNA-GAS5 promotes spinal cord repair and the inhibition of neuronal apoptosis via the transplantation of 3D printed scaffold loaded with induced pluripotent stem cell-derived neural stem cells. Ann. Transl. Med. 2021, 9, 931. [Google Scholar] [CrossRef]
  177. Zarepour, A.; Öztürk, A.B.; Irmak, D.K.; Yaşayan, G.; Gökmen, A.; Karaöz, E.; Zarepour, A.; Zarrabi, A.; Mostafavi, E. Combination Therapy Using Nanomaterials and Stem Cells to Treat Spinal Cord Injuries. Eur. J. Pharm. Biopharm. 2022, 177, 224–240. [Google Scholar] [CrossRef]
  178. Nejati, K.; Mehdi, D.; Ghareghomi, S.; Mostafavi, E.; Ebrahimi-Kalan, A.; Biglari, A.; Alizadeh, E.; Mortazavi, Y.; Zarghami, N. GDNF gene-engineered adipose-derived stem cells seeded Emu oil-loaded electrospun nanofibers for axonal regeneration following spinal cord injury. J. Drug Deliv. Sci. Technol. 2020, 60, 102095. [Google Scholar] [CrossRef]
  179. Lv, B.; Zhang, X.; Yuan, J.; Chen, Y.; Ding, H.; Cao, X.; Huang, A. Biomaterial-supported MSC transplantation enhances cell–cell communication for spinal cord injury. Stem Cell Res. Ther. 2021, 12, 36. [Google Scholar] [CrossRef]
  180. Papa, S.; Vismara, I.; Mariani, A.; Barilani, M.; Rimondo, S.; De Paola, M.; Panini, N.; Erba, E.; Mauri, E.; Rossi, F.; et al. Mesenchymal stem cells encapsulated into biomimetic hydrogel scaffold gradually release CCL2 chemokine in situ preserving cytoarchitecture and promoting functional recovery in spinal cord injury. J. Control. Release 2018, 278, 49–56. [Google Scholar] [CrossRef] [PubMed]
  181. Park, S.-S.; Lee, Y.J.; Lee, S.H.; Lee, D.; Choi, K.; Kim, W.-H.; Kweon, O.-K.; Han, H.J. Functional recovery after spinal cord injury in dogs treated with a combination of Matrigel and neural-induced adipose-derived mesenchymal Stem cells. Cytotherapy 2012, 14, 584–597. [Google Scholar] [CrossRef]
  182. Raynald, S.B.; Liu, X.-B.; Zhou, J.F.; Huang, H.; Wang, J.-Y.; Sun, X.-D.; Qin, C.; An, Y.-H. Polypyrrole/polylactic acid nanofibrous scaffold cotransplanted with bone marrow stromal cells promotes the functional recovery of spinal cord injury in rats. CNS Neurosci. Ther. 2019, 25, 951–964. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, C.; Xu, H.-H.; Liu, X.-Y.; Zhang, Y.-S.; Zhong, L.; Wang, Y.-W.; Xu, L.; Wei, P.; Chen, Y.-X.; Liu, P.; et al. 3D printed collagen/silk fibroin scaffolds carrying the secretome of human umbilical mesenchymal stem cells ameliorated neurological dysfunction after spinal cord injury in rats. Regen. Biomater. 2022, 9, rbac014. [Google Scholar] [CrossRef]
  184. Zaviskova, K.; Tukmachev, D.; Dubisova, J.; Vackova, I.; Hejcl, A.; Bystronova, J.; Pravda, M.; Scigalkova, I.; Sulakova, R.; Velebny, V.; et al. Injectable hydroxyphenyl derivative of hyaluronic acid hydrogel modified with RGD as scaffold for spinal cord injury repair. J. Biomed. Mater. Res. Part A 2018, 106, 1129–1140. [Google Scholar] [CrossRef] [PubMed]
  185. Jiao, G.; Lou, G.; Mo, Y.; Pan, Y.; Zhang, Z.; Guo, R.; Li, Z. A combination of GDNF and hUCMSC transplantation loaded on SF/AGs composite scaffolds for spinal cord injury repair. Mater. Sci. Eng. C 2017, 74, 230–237. [Google Scholar] [CrossRef]
  186. Cholas, R.; Hsu, H.P.; Spector, M. Collagen Scaffolds Incorporating Select Therapeutic Agents to Facilitate a Reparative Response in a Standardized Hemiresection Defect in the Rat Spinal Cord. Tissue Eng. Part A 2012, 18, 2158–2172. [Google Scholar] [CrossRef]
  187. Terraf, P.; Kouhsari, S.M.; Ai, J.; Babaloo, H. Tissue-Engineered Regeneration of Hemisected Spinal Cord Using Human Endometrial Stem Cells, Poly ε-Caprolactone Scaffolds, and Crocin as a Neuroprotective Agent. Mol. Neurobiol. 2017, 54, 5657–5667. [Google Scholar] [CrossRef]
  188. Günther, M.I.; Weidner, N.; Müller, R.; Blesch, A. Cell-seeded alginate hydrogel scaffolds promote directed linear axonal regeneration in the injured rat spinal cord. Acta Biomater. 2015, 27, 140–150. [Google Scholar] [CrossRef]
  189. Zhang, J.; Cheng, T.; Chen, Y.; Gao, F.; Guan, F.; Yao, M.-H. A chitosan-based thermosensitive scaffold loaded with bone marrow-derived mesenchymal stem cells promotes motor function recovery in spinal cord injured mice. Biomed. Mater. 2020, 15, 035020. [Google Scholar] [CrossRef]
  190. Li, Y.; Yang, L.; Hu, F.; Xu, J.; Ye, J.; Liu, S.; Wang, L.; Zhuo, M.; Ran, B.; Zhang, H.; et al. Novel Thermosensitive Hydrogel Promotes Spinal Cord Repair by Regulating Mitochondrial Function. ACS Appl. Mater. Interfaces 2022, 14, 25155–25172. [Google Scholar] [CrossRef]
  191. Blaško, J.; Szekiova, E.; Slovinska, L.; Kafka, J.; Cizkova, D. Axonal outgrowth stimulation after alginate/mesenchymal stem cell therapy in injured rat spinal cord. Acta Neurobiol. Exp. 2017, 77, 337–350. [Google Scholar] [CrossRef] [Green Version]
  192. Zurita, M.; Otero, L.; Aguayo, C.; Bonilla, C.; Ferreira, E.; Parajón, A.; Vaquero, J. Cell therapy for spinal cord repair: Optimization of biologic scaffolds for survival and neural differentiation of human bone marrow stromal cells. Cytotherapy 2010, 12, 522–537. [Google Scholar] [CrossRef] [PubMed]
  193. An, H.; Li, Q.; Wen, J. Bone marrow mesenchymal stem cells encapsulated thermal-responsive hydrogel network bridges combined photo-plasmonic nanoparticulate system for the treatment of urinary bladder dysfunction after spinal cord injury. J. Photochem. Photobiol. B Biol. 2019, 203, 111741. [Google Scholar] [CrossRef]
  194. Chen, X.; Yang, Y.; Yao, J.; Lin, W.; Li, Y.; Chen, Y.; Gao, Y.; Yang, Y.; Gu, X.; Wang, X. Bone marrow stromal cells-loaded chitosan conduits promote repair of complete transection injury in rat spinal cord. J. Mater. Sci. Mater. Med. 2011, 22, 2347–2356. [Google Scholar] [CrossRef] [PubMed]
  195. Huang, C.; Liu, Y.; Ding, J.; Dai, Y.; Le, L.; Wang, L.; Ding, E.; Yang, J. Thermosensitive quaternized chitosan hydrogel scaffolds promote neural differentiation in bone marrow mesenchymal stem cells and functional recovery in a rat spinal cord injury model. Cell Tissue Res. 2021, 385, 65–85. [Google Scholar] [CrossRef] [PubMed]
  196. Liu, D.; Li, X.; Xiao, Z.; Yin, W.; Zhao, Y.; Tan, J.; Chen, B.; Jiang, X.; Dai, J. Different functional bio-scaffolds share similar neurological mechanism to promote locomotor recovery of canines with complete spinal cord injury. Biomaterials 2019, 214, 119230. [Google Scholar] [CrossRef]
  197. Wiseman, T.M.; Baron-Heeris, M.D.; Houwers, I.G.J.; Keenan, R.; Williams, R.J.; Nisbet, D.R.; Harvey, A.R.; Hodgetts, S.I. Peptide Hydrogel Scaffold for Mesenchymal Precursor Cells Implanted to Injured Adult Rat Spinal Cord. Tissue Eng. Part A 2021, 27, 993–1007. [Google Scholar] [CrossRef]
  198. Wang, X.-H.; Tang, X.-C.; Li, X.; Qin, J.-Z.; Zhong, W.-T.; Wu, P.; Zhang, F.; Shen, Y.-X.; Dai, T.-T. Implantation of nanofibrous silk scaffolds seeded with bone marrow stromal cells promotes spinal cord regeneration (6686 words). Artif. Cells, Nanomed. Biotechnol. 2021, 49, 699–708. [Google Scholar] [CrossRef]
  199. Han, I.-B.; Thakor, D.K.; Ropper, A.E.; Yu, D.; Wang, L.; Kabatas, S.; Zeng, X.; Kim, S.-W.; Zafonte, R.D.; Teng, Y.D. Physical impacts of PLGA scaffolding on hMSCs: Recovery neurobiology insight for implant design to treat spinal cord injury. Exp. Neurol. 2019, 320, 112980. [Google Scholar] [CrossRef]
  200. Kang, K.N.; Kim, D.Y.; Yoon, S.M.; Lee, J.Y.; Lee, B.N.; Kwon, J.S.; Seo, H.W.; Lee, I.W.; Shin, H.C.; Kim, Y.M.; et al. Tissue engineered regeneration of completely transected spinal cord using human mesenchymal stem cells. Biomaterials 2012, 33, 4828–4835. [Google Scholar] [CrossRef]
  201. Kang, K.N.; Lee, J.Y.; Kim, D.Y.; Lee, B.N.; Ahn, H.H.; Lee, B.; Khang, G.; Park, S.R.; Min, B.H.; Kim, J.H.; et al. Regeneration of Completely Transected Spinal Cord Using Scaffold of Poly(D,L-Lactide-co-Glycolide)/Small Intestinal Submucosa Seeded with Rat Bone Marrow Stem Cells. Tissue Eng. Part A 2011, 17, 2143–2152. [Google Scholar] [CrossRef] [PubMed]
  202. Peng, Z.; Gao, W.; Yue, B.; Jiang, J.; Gu, Y.; Dai, J.; Chen, L.; Shi, Q. Promotion of neurological recovery in rat spinal cord injury by mesenchymal stem cells loaded on nerve-guided collagen scaffold through increasing alternatively activated macrophage polarization. J. Tissue Eng. Regen. Med. 2018, 12, e1725–e1736. [Google Scholar] [CrossRef] [PubMed]
  203. Yuan, X.; Yuan, W.; Ding, L.; Shi, M.; Luo, L.; Wan, Y.; Oh, J.; Zhou, Y.; Bian, L.; Deng, D.Y.B. Cell-adaptable dynamic hydrogel reinforced with stem cells improves the functional repair of spinal cord injury by alleviating neuroinflammation. Biomaterials 2021, 279, 121190. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, L.; Wan, J.; Dai, M.; Ye, X.; Liu, C.; Tang, C.; Zhu, L. Effects of oxygen generating scaffolds on cell survival and functional recovery following acute spinal cord injury in rats. J. Mater. Sci. Mater. Med. 2020, 31, 115. [Google Scholar] [CrossRef] [PubMed]
  205. Ropper, A.E.; Thakor, D.K.; Han, I.; Yu, D.; Zeng, X.; Anderson, J.E.; Aljuboori, Z.; Kim, S.-W.; Wang, H.; Sidman, R.L.; et al. Defining recovery neurobiology of injured spinal cord by synthetic matrix-assisted hMSC implantation. Proc. Natl. Acad. Sci. USA 2017, 114, E820–E829. [Google Scholar] [CrossRef] [Green Version]
  206. Li, X.; Tan, J.; Xiao, Z.; Zhao, Y.; Han, S.; Liu, D.; Yin, W.; Li, J.; Li, J.; Wanggou, S.; et al. Transplantation of hUC-MSCs seeded collagen scaffolds reduces scar formation and promotes functional recovery in canines with chronic spinal cord injury. Sci. Rep. 2017, 7, 43559. [Google Scholar] [CrossRef] [Green Version]
  207. Li, L.; Xiao, B.; Mu, J.; Zhang, Y.; Zhang, C.; Cao, H.; Chen, R.; Patra, H.K.; Yang, B.; Feng, S.; et al. A MnO2 Nanoparticle-Dotted Hydrogel Promotes Spinal Cord Repair via Regulating Reactive Oxygen Species Microenvironment and Synergizing with Mesenchymal Stem Cells. ACS Nano 2019, 13, 14283–14293. [Google Scholar] [CrossRef]
  208. Li, Y.; Cao, X.; Deng, W.; Yu, Q.; Sun, C.; Ma, P.; Shao, F.; Yusif, M.M.; Ge, Z.; Wang, K.; et al. 3D printable Sodium alginate-Matrigel (SA-MA) hydrogel facilitated ectomesenchymal stem cells (EMSCs) neuron differentiation. J. Biomater. Appl. 2021, 35, 709–719. [Google Scholar] [CrossRef] [PubMed]
  209. Monje, P.V.; Deng, L.; Xu, X.-M. Human Schwann Cell Transplantation for Spinal Cord Injury: Prospects and Challenges in Translational Medicine. Front. Cell. Neurosci. 2021, 15, 690894. [Google Scholar] [CrossRef] [PubMed]
  210. Hakim, J.S.; Rad, M.E.; Grahn, P.J.; Chen, B.K.; Knight, A.M.; Schmeichel, A.M.; Isaq, N.A.; Dadsetan, M.; Yaszemski, M.J.; Windebank, A.J. Positively Charged Oligo[Poly(Ethylene Glycol) Fumarate] Scaffold Implantation Results in a Permissive Lesion Environment after Spinal Cord Injury in Rat. Tissue Eng. Part A 2015, 21, 2099–2114. [Google Scholar] [CrossRef] [Green Version]
  211. Hurtado, A.; Moon, L.D.; Maquet, V.; Blits, B.; Jérôme, R.; Oudega, M. Poly (d,l-lactic acid) macroporous guidance scaffolds seeded with Schwann cells genetically modified to secrete a bi-functional neurotrophin implanted in the completely transected adult rat thoracic spinal cord. Biomaterials 2006, 27, 430–442. [Google Scholar] [CrossRef] [Green Version]
  212. Liu, S.; Sandner, B.; Schackel, T.; Nicholson, L.; Chtarto, A.; Tenenbaum, L.; Puttagunta, R.; Müller, R.; Weidner, N.; Blesch, A. Regulated viral BDNF delivery in combination with Schwann cells promotes axonal regeneration through capillary alginate hydrogels after spinal cord injury. Acta Biomater. 2017, 60, 167–180. [Google Scholar] [CrossRef] [Green Version]
  213. Moore, M.J.; Friedman, J.A.; Lewellyn, E.B.; Mantila, S.M.; Krych, A.J.; Ameenuddin, S.; Knight, A.M.; Lu, L.; Currier, B.L.; Spinner, R.J.; et al. Multiple-channel scaffolds to promote spinal cord axon regeneration. Biomaterials 2006, 27, 419–429. [Google Scholar] [CrossRef] [PubMed]
  214. Oudega, M.; Gautier, S.E.; Chapon, P.; Fragoso, M.; Bates, M.L.; Parel, J.M.; Bunge, M.B. Axonal regeneration into Schwann cell grafts within resorbable poly(α-hydroxyacid) guidance channels in the adult rat spinal cord. Biomaterials 2001, 22, 1125–1136. [Google Scholar] [CrossRef] [PubMed]
  215. Chen, B.K.; Knight, A.M.; de Ruiter, G.C.; Spinner, R.J.; Yaszemski, M.J.; Currier, B.L.; Windebank, A.J. Axon regeneration through scaffold into distal spinal cord after transection. J. Neurotrauma 2009, 6, 1759–1771. [Google Scholar] [CrossRef] [PubMed]
  216. Novikov, L.N.; Novikova, L.N.; Mosahebi, A.; Wiberg, M.; Terenghi, G.; Kellerth, J.-O. A novel biodegradable implant for neuronal rescue and regeneration after spinal cord injury. Biomaterials 2002, 23, 3369–3376. [Google Scholar] [CrossRef]
  217. Novikova, L.N.; Pettersson, J.; Brohlin, M.; Wiberg, M.; Novikov, L.N. Biodegradable poly-β-hydroxybutyrate scaffold seeded with Schwann cells to promote spinal cord repair. Biomaterials 2008, 29, 1198–1206. [Google Scholar] [CrossRef] [PubMed]
  218. Babaloo, H.; Ebrahimi-Barough, S.; Derakhshan, M.A.; Yazdankhah, M.; Lotfibakhshaiesh, N.; Soleimani, M.; Joghataei, M.-T.; Ai, J. PCL/gelatin nanofibrous scaffolds with human endometrial stem cells/Schwann cells facilitate axon regeneration in spinal cord injury. J. Cell. Physiol. 2019, 234, 11060–11069. [Google Scholar] [CrossRef] [PubMed]
  219. Yang, E.-Z.; Zhang, G.-W.; Xu, J.-G.; Chen, S.; Wang, H.; Cao, L.-L.; Liang, B.; Lian, X.-F. Multichannel polymer scaffold seeded with activated Schwann cells and bone mesenchymal stem cells improves axonal regeneration and functional recovery after rat spinal cord injury. Acta Pharmacol. Sin. 2017, 38, 623–637. [Google Scholar] [CrossRef] [Green Version]
  220. Zhang, J.; Lu, X.; Feng, G.; Gu, Z.; Sun, Y.; Bao, G.; Xu, G.; Lu, Y.; Chen, J.; Xu, L.; et al. Chitosan scaffolds induce human dental pulp stem cells to neural differentiation: Potential roles for spinal cord injury therapy. Cell Tissue Res. 2016, 366, 129–142. [Google Scholar] [CrossRef]
  221. Li, X.; Yang, C.; Li, L.; Xiong, J.; Xie, L.; Yang, B.; Yu, M.; Feng, L.; Jiang, Z.; Guo, W.; et al. A Therapeutic Strategy for Spinal Cord Defect: Human Dental Follicle Cells Combined with Aligned PCL/PLGA Electrospun Material. BioMed Res. Int. 2015, 2015, 197183. [Google Scholar] [CrossRef] [Green Version]
  222. Thompson, R.E.; Pardieck, J.; Smith, L.; Kenny, P.; Crawford, L.; Shoichet, M.; Sakiyama-Elbert, S. Effect of hyaluronic acid hydrogels containing astrocyte-derived extracellular matrix and/or V2a interneurons on histologic outcomes following spinal cord injury. Biomaterials 2018, 162, 208–223. [Google Scholar] [CrossRef]
  223. Wang, C.; Sun, C.; Hu, Z.; Huo, X.; Yang, Y.; Liu, X.; Botchway, B.O.A.; Davies, H.; Fang, M. Improved Neural Regeneration with Olfactory Ensheathing Cell Inoculated PLGA Scaffolds in Spinal Cord Injury Adult Rats. Neurosignals 2017, 25, 1–14. [Google Scholar] [CrossRef] [Green Version]
  224. Ferrero-Gutierrez, A.; Menendez-Menendez, Y.; Alvarez-Viejo, M.; Meana, A.; Otero, J. New serum-derived albumin scaffold seeded with adipose-derived stem cells and olfactory ensheathing cells used to treat spinal cord injured rats. Histol. Histopathol. 2013, 28, 89–100. [Google Scholar] [CrossRef]
  225. Wang, N.; Xiao, Z.; Zhao, Y.; Wang, B.; Li, X.; Li, J.; Dai, J. Collagen scaffold combined with human umbilical cord-derived mesenchymal stem cells promote functional recovery after scar resection in rats with chronic spinal cord injury. J. Tissue Eng. Regen. Med. 2018, 12, e1154–e1163. [Google Scholar] [CrossRef]
  226. Amr, S.M.; Gouda, A.; Koptan, W.T.; Galal, A.A.; Abdel-Fattah, D.S.; Rashed, L.A.; Atta, H.M.; Abdel-Aziz, M.T. Bridging defects in chronic spinal cord injury using peripheral nerve grafts combined with a chitosan-laminin scaffold and enhancing regeneration through them by co-transplantation with bone-marrow-derived mesenchymal stem cells: Case series of 14 patients. J. Spinal Cord Med. 2014, 37, 54–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Tian, L.; Prabhakaran, M.P.; Ramakrishna, S. Strategies for regeneration of components of nervous system: Scaffolds, cells and biomolecules. Regen. Biomater. 2015, 2, 31–45. [Google Scholar] [CrossRef] [Green Version]
  228. Raspa, A.; Marchini, A.; Pugliese, R.; Mauri, M.; Maleki, M.; Vasita, R.; Gelain, F. A biocompatibility study of new nanofibrous scaffolds for nervous system regeneration. Nanoscale 2016, 8, 253–265. [Google Scholar] [CrossRef] [PubMed]
  229. Li, X.; Liu, D.; Xiao, Z.; Zhao, Y.; Han, S.; Chen, B.; Dai, J. Scaffold-facilitated locomotor improvement post complete spinal cord injury: Motor axon regeneration versus endogenous neuronal relay formation. Biomaterials 2019, 197, 20–31. [Google Scholar] [CrossRef] [PubMed]
  230. Domínguez-Bajo, A.; González-Mayorga, A.; Guerrero, C.R.; Palomares, F.J.; García, R.; López-Dolado, E.; Serrano, M.C. Myelinated axons and functional blood vessels populate mechanically compliant rGO foams in chronic cervical hemisected rats. Biomaterials 2019, 192, 461–474. [Google Scholar] [CrossRef] [PubMed]
  231. Austin, J.W.; Kang, C.E.; Baumann, M.D.; DiDiodato, L.; Satkunendrarajah, K.; Wilson, J.R.; Stanisz, G.J.; Shoichet, M.S.; Fehlings, M.G. The effects of intrathecal injection of a hyaluronan-based hydrogel on inflammation, scarring and neurobehavioural outcomes in a rat model of severe spinal cord injury associated with arachnoiditis. Biomaterials 2012, 33, 4555–4564. [Google Scholar] [CrossRef] [PubMed]
  232. Huang, L.; Wang, Y.; Zhu, M.; Wan, X.; Zhang, H.; Lei, T.; Blesch, A.; Liu, S. Anisotropic Alginate Hydrogels Promote Axonal Growth across Chronic Spinal Cord Transections after Scar Removal. ACS Biomater. Sci. Eng. 2020, 6, 2274–2286. [Google Scholar] [CrossRef] [PubMed]
  233. Tavakol, S.; Saber, R.; Hoveizi, E.; Aligholi, H.; Ai, J.; Rezayat, S.M. Chimeric Self-assembling Nanofiber Containing Bone Marrow Homing Peptide’s Motif Induces Motor Neuron Recovery in Animal Model of Chronic Spinal Cord Injury; an In Vitro and In Vivo Investigation. Mol. Neurobiol. 2016, 53, 3298–3308. [Google Scholar] [CrossRef] [PubMed]
  234. Nguyen, L.H.; Gao, M.; Lin, J.; Wu, W.; Wang, J.; Chew, S.Y. Three-dimensional aligned nanofibers-hydrogel scaffold for controlled non-viral drug/gene delivery to direct axon regeneration in spinal cord injury treatment. Sci. Rep. 2017, 7, srep42212, Erratum in Sci. Rep. 2018, 8, 13057. [Google Scholar] [CrossRef] [PubMed]
  235. López-Dolado, E.; González-Mayorga, A.; Gutiérrez, M.C.; Serrano, M.C. Immunomodulatory and angiogenic responses induced by graphene oxide scaffolds in chronic spinal hemisected rats. Biomaterials 2016, 99, 72–81. [Google Scholar] [CrossRef] [PubMed]
  236. Gelain, F.; Panseri, S.; Antonini, S.; Cunha, C.; Donega, M.; Lowery, J.; Taraballi, F.; Cerri, G.; Montagna, M.; Baldissera, F.; et al. Transplantation of Nanostructured Composite Scaffolds Results in the Regeneration of Chronically Injured Spinal Cords. ACS Nano 2011, 5, 227–236. [Google Scholar] [CrossRef] [PubMed]
  237. Xiao, Z.; Tang, F.; Zhao, Y.; Han, G.; Yin, N.; Li, X.; Chen, B.; Han, S.; Jiang, X.; Yun, C.; et al. Significant Improvement of Acute Complete Spinal Cord Injury Patients Diagnosed by a Combined Criteria Implanted with NeuroRegen Scaffolds and Mesenchymal Stem Cells. Cell Transplant. 2018, 27, 907–915. [Google Scholar] [CrossRef]
Figure 1. The diagram shows the pathophysiological events in SCI. Progressive demyelination results in the degeneration of axonal fibers. A cavitation occurs in the epicenter. Hypertrophic astrocytes with very long processes over the tips of non-regenerating fibers form a barrier known as a glial wall around the cavitation. In response to injury, microglial cells transform into active phagocytic microglia and exhibit chemotaxis. The presence of CSPGs creates an inhibitory environment for axonal regeneration. In addition, CSPG also inhibits the migration and differentiation of oligodendrocyte progenitor cells.
Figure 1. The diagram shows the pathophysiological events in SCI. Progressive demyelination results in the degeneration of axonal fibers. A cavitation occurs in the epicenter. Hypertrophic astrocytes with very long processes over the tips of non-regenerating fibers form a barrier known as a glial wall around the cavitation. In response to injury, microglial cells transform into active phagocytic microglia and exhibit chemotaxis. The presence of CSPGs creates an inhibitory environment for axonal regeneration. In addition, CSPG also inhibits the migration and differentiation of oligodendrocyte progenitor cells.
Ijms 24 02528 g001
Figure 2. The diagram shows the pathophysiological change following a biomaterial scaffold graft. Certain biomaterials contain stem cells, drugs, neurotrophines or growth factors, etc. Grafted biomaterials support axonal regeneration beyond the glial scar as a scaffold. Grafted biomaterial scaffolds release the transplanted cells or secreted neurotrophines, and, in addition, they release the encapsulated drugs or growth factors, etc. They support the formation of new synaptic circuits and connectivity between host neurons and axons, and, in addition, they improve morphological and behavioral outcomes after experimental SCI. Oligodendrocytes derived from grafted stem cells remyelinate damaged host axons. Regenerated and remyelinated axons pass through the injured lesion and connect to other host neurons supported by interneurons and glial cells derived from grafted stem cells.
Figure 2. The diagram shows the pathophysiological change following a biomaterial scaffold graft. Certain biomaterials contain stem cells, drugs, neurotrophines or growth factors, etc. Grafted biomaterials support axonal regeneration beyond the glial scar as a scaffold. Grafted biomaterial scaffolds release the transplanted cells or secreted neurotrophines, and, in addition, they release the encapsulated drugs or growth factors, etc. They support the formation of new synaptic circuits and connectivity between host neurons and axons, and, in addition, they improve morphological and behavioral outcomes after experimental SCI. Oligodendrocytes derived from grafted stem cells remyelinate damaged host axons. Regenerated and remyelinated axons pass through the injured lesion and connect to other host neurons supported by interneurons and glial cells derived from grafted stem cells.
Ijms 24 02528 g002
Figure 3. Flowchart of the screening process in this systematic review.
Figure 3. Flowchart of the screening process in this systematic review.
Ijms 24 02528 g003
Table 1. Summary of the included studies and the effect of hydrogel application in SCI.
Table 1. Summary of the included studies and the effect of hydrogel application in SCI.
Effect on Pathophysiological Events
Author, YearLocation of InjurySpeciesApplicationHydrogel (Character)Anti-InflammationScar/CavityAxon GrowthAngiogenesisMotor Functional Recovery
Sun Y, et al.,
2019 [33]
ThoracicRatImplantCollagen/Chitosan
(3D printing)
NA++NA+
Marchand R, et al.,
1990 [34]
ThoracicRatImplantCollagen
(Self assembling)
NA++NANA
Khan T, et al.,
1991 [35]
ThoracicRatImplantCarbon
(Filament)
NA++NANA
Liu W, et al.,
2020 [36]
ThoracicRatImplantCollagen
(modified with N-cadherin)
NA++NA+
Fan C, et al.,
2017 [37]
ThoracicRatImplantCollagen
(binding with EGFR antibody Fab fragment)
NA++NA+
Yang B, et al.,
2017 [38]
ThoracicRatImplantAgarose/gelatin/polypyrrole
(similar conductivity as the spinal cord)
+++NA+
Martín-López E, et al.,
2013 [39]
ThoracicRatImplantAgarose with
κ-carrageenan,
gelatin, xanthan gum and polysulfone
NA++NANA
Gros T, et al.,
2010 [40]
CervicalRatImplantAgaroseNA++NANA
Kataoka K, et al.,
2004 [41]
ThoracicRatImplantAlginate
(Freeze-dried sponge )
NA++NANA
Prang P, et al.,
2006 [42]
CervicalRatImplantAlginate
(Anisotropic capillary)
NA++NANA
Cao Z, et al.,
2020 [43]
LumbarCanineImplantFibrin
(Hierarchically aligned fibrin hydrogel)
NA++NA+
Yin W, et al.,
2021 [44]
ThoracicCanineImplantCollagen
(Taxol-modified linear-ordered scaffold)
NA++NANA
Altinova H, et al.,
2020 [45]
CervicalRatImplantCollagen NA+++NA
Gholami M, et al.,
2021 [46]
ThoracicRatImplant Chitosan/alginate/erythropoietin +++NANA
Stokols S, et al.,
2006 [47]
CervicalRatImplantAlginate
(recombinant BDNF protein)
NA++NANA
Zhang Z, et al.,
2017 [48]
LumbarCanineImplantBiomaterial-aligned fibrin+++NA+
Fukushima K, et al.,
2008 [49]
ThoracicRatImplantCollagen
(Honeycomb)
NANA+NANA
Zhao X, et al.,
2022 [50]
ThoracicRatImplantGelatin/hyaluronic acid +++NA+
King VR, et al.,
2010 [51]
ThoracicRatImplantCollagen
(viscous fibronectin gel)
NANA+NANA
Cheng H, et al.,
2007 [52]
ThoracicRatImplant Chitosan NA++NA-
Han S, et al.,
2018 [53]
ThoracicRatImplantAgarose
(Matrigel)
NANA+NA+
Bakshi A, et al.,
2004 [54]
CervicalRatImplantNonbiodegradable hydrogel
(pPHEMA)
++++NA
Zhai H, et al.,
2020 [55]
ThoracicRatImplantADA16 peptide hydrogelNA+++NA
Hejčl A, et al.,
2018 [56]
ThoracicRatImplant3 Methacrylate hydrogelNA++++
Zhang Q, et al.,
2016 [57]
ThoracicRatImplantSilk protein/lamininNA++++
Chai Y, et al.,
2022 [58]
ThoracicRatImplantBioactive isoleucine-lysine-valine-alanine-valineNA++++
Yang Y, et al.,
2021 [60]
ThoracicRatImplantInjectable collagen hydrogelNA++NA+
Silva NA, et al.,
2010 [59]
ThoracicRatImplantStarch/poly-e-caprolactone
blend and gellan gum
+NANANANA
Suzuki H, et al.,
2015 [61]
ThoracicRatImplantCollagen filamentsNA++NANA
Yara T, et al.,
2009 [62]
ThoracicRatImplantCollagen filamentsNA++NANA
Note: -, no difference with the control group; +, effective; NA, not available.
Table 2. Summary of the included studies and the effect of biodegradable scaffolds application in SCI.
Table 2. Summary of the included studies and the effect of biodegradable scaffolds application in SCI.
Effect On Pathophysiological Events
Author, YearLocation of InjurySpeciesApplicationBiodegradable Scaffold/(Character)Anti-InflammationScar/CavityAxon GrowthAngiogenesisMotor Functional Recovery
Kubinová Š, et al.,
2015 [66]
ThoracicRatImplantSIKVAV-modified PHEMANA+++NA
Hejcl A, et al.,
2008 [67]
ThoracicRatImplant2-hydroxyethyl methacrylateNA+++NA
Slotkin JR, et al.,
2017 [69]
ThoracicThe green monkeyImplantPoly-lactic-co-glycolic acid and Poly-l-lysine+++NANA
Silva NA, et al.,
2013 [70]
ThoracicRatImplantStarch with polycaprolactoneNA++NA+
Thomas AM, et al.,
2013 [71]
ThoracicRat/MouseImplantPoly(lactide-co-glycolide)
multiple channel bridges
NA++NANA
Man W, et al.,
2021 [72]
ThoracicRatImplantHierarchically aligned fibrin hydrogel
and functionalized self-assembling peptides
NA++++
Kubinová S, et al.,
2011 [73]
ThoracicRatImplantHighly superporous cholesterol-modified
poly(2-hydroxyethyl
methacrylate) scaffolds
NA+++NA
Guest JD, et al.,
2018 [74]
ThoracicThoracicImplantPLGA-PLLNA++NA-
Hakim JS, et al.,
2019 [75]
ThoracicRatImplantPLGA-PLLNA++NA-
Anzalone A, et al.,
2018 [76]
CervicalMouseImplantPoly-lactic-co-glycolicNANA+NANA
De Laporte L, et al.,
2009 [77]
ThoracicRatImplantPoly-lactic-co-glycolic/(Lipoplex incubation on ECM-coated PLG)NANANANANA
Wong DY, et al.,
2008 [78]
ThoracicRatImplantSalt-leached porous poly
(epsilon-caprolactone)
NANA+NANA
Ribeiro-Samy S, et al.,
2013 [79]
ThoracicRatImplantPoly(3-hydroxybutyrateco-
3-hydroxyvalerate) (PHB-HV)
NANANANA-
Pawar K, et al.,
2015 [80]
CervicalMouseImplant Poly-lactic-co-glycolicNANA+NA+
Rooney GE, et al.,
2008 [81]
ThoracicRatImplantRadiopaque barium sulfate-impregnated poly-lactic-co-glycolic acidNANANANANA
Shu B, et al.,
2019 [82]
ThoracicRatImplant PLA-PPy +++NA+
Zhou L, et al.,
2018 [83]
ThoracicMouseImplantPlant-derived polyphenol, tannic acid (TA), cross-linking and doping
conducting polypyrrole (PPy) chains
NA++NA+
Pertici VA, et al.,
2014 [84]
ThoracicRatImplantPLA-b-PHEMA block
copolymer
+++++
Reis KP, et al.,
2020 [85]
ThoracicRatImplantValproic acid (VPA)/PLGA
(Microfiber)
+++++
Novikova LN, et al.,
2017 [86]
CervicalRatImplantTrimethylene carbonate and
e-caprolactone (TC)
containing poly-p-dioxanone
microfilaments (PDO)
-++NANA
Note: -, no difference with the control group; +, effective; NA, not available.
Table 3. Summary of the included studies and the effect of nano-/micro-scale biomaterial scaffolds application in SCI.
Table 3. Summary of the included studies and the effect of nano-/micro-scale biomaterial scaffolds application in SCI.
Effect on Pathophysiological Events
Author, YearLocation of InjurySpeciesApplicationNanomaterial Scaffold/MaterialAnti-InflammationScar/CavityAxon GrowthAngiogenesisMotor Functional Recovery
Zamani F, et al.,
2014 [89]
ThoracicRatImplant3D nanofibrous core–sheath scaffold/PLGANANA+++
Sun X, et al.,
2019 [90]
ThoracicRatImplantNano-fibrous channel wall/PLLA+++NA+
Cigognini D, et al.,
2014 [91]
ThoracicRatInjectedNanostructures of two self-assembling peptides B24 and biotin-LDLK12NANA+NANA
Yao S, et al.,
2018 [92]
ThoracicRatImplantHierarchically aligned fibrin nanofiber/Fibrin hydrogelNA++++
Altinova H, et al.,
2016 [93]
CervicalRatImplantMicrostructured scaffold/Collagen+++++
Usmani S, et al.,
2020 [94]
ThoracicRatImplantArtificial nanotube/Carbon+++NA+
Sever-Bahcekapili M, et al.,
2020 [95]
ThoracicRatImplantNeuroactive peptide nanofibers/
LN-PA, GAG-PA
NA++NA+
Zhao T, et al.,
2018 [96]
ThoracicRatImplantNanofibrous scaffolds/
PHBV, PLA, Collagen
NA++NA-
Chedly JL, et al.,
2017 [97]
ThoracicRatImplantMicrohydrogel scaffold/Chitosan+++++
Cigognini D, et al.,
2011 [98]
ThoracicRatImplantNanomaterial SAPs with bone marrow homing motif (BMHP1)+++++
Palejwala AH, et al.,
2016 [99]
ThoracicRatImplantPoly (3-hydroxybutyrateco-
3-hydroxyvalerate) (PHB-HV)
NANANANA-
Palejwala AH, et al.,
2016 [99]
ThoracicRatImplantNanoscaffoldsNA+++NA
Pawelec KM, et al.,
2018 [100]
ThoracicRatImplantMicrostructure multi-channel scaffold/PCLNANA+NANA
Milbreta U, 2016 [101]CervicalRatImplant 3D nanofiber scaffold/Collagen +++NANA
Tysseling VM, et al.,
2010 [102]
ThoracicRatInjectedPeptide amphiphile (PA) molecules that self-assemble and display the laminin epitope IKVAVNA++NA+
Liu Y, et al.,
2013 [103]
ThoracicRatInjectedA self-assembling peptide/
K2(QL)6K2 (QL6)
+++NA+
Note: -, no difference with the control group; +, effective; NA, not available.
Table 4. Summary of the included studies and the effect of biomaterial scaffolds in combinatory treatment for SCI.
Table 4. Summary of the included studies and the effect of biomaterial scaffolds in combinatory treatment for SCI.
Effect on Pathophysiological Events
Author, YearLocation of InjurySpeciesCombinatory AgentBiomaterial Scaffold Anti-InflammationScar/CavityAxon GrowthAngiogenesisFacilitation of Cell MigrationMotor Functional Recovery
Furuya T, et al.,
2013 [105]
ThoracicRatbFGFGelatin hydrogelNANANANANANA
Chantal SA, et al.,
2008 [106]
ThoracicRatMethylprednisolone Biodegradable PLGA-based nanoparticles ++NANA+NA
Jain A, et al.,
2011 [108]
ThoracicRatConstitutively active Cdc42,
Rac1, BDNF
Microtubule-mediated slow release of BDNF+++NA+NA
Wen Y et al.,
2016 [109]
ThoracicRatAnti-Nogo receptor antibodyPLGA microspheres
containing BDNF and VEGF
++++++
Chen B, et al.,
2015 [110]
ThoracicRatbFGFHEMA-MOETACL hydrogelNA++NANA+
Lin J, et al.,
2019 [114]
ThoracicRatRehabilitationHybrid fiber-hydrogel scaffold+++NA++
Shi Q, et al.,
2014 [115]
ThoracicRatbFGFCollagen scaffoldNA++NA++
Wang X, et al.,
2013 [116]
ThoracicRatNT-3Chitosan-based tube scaffoldNA++NA++
Li G, et al.,
2016 [117]
ThoracicRat and canineNT-3Fibrin-coated gelatin sponge scaffold+++NA++
Wei YT, et al.,
2010 [118]
ThoracicRatNogo-66 receptor antibodyHyaluronic acid -based hydrogels modified with poly-L-lysine (PLL) +++++NA
Bighinati A, et al.,
2020 [119]
ThoracicRatIbuprofen and triiodothyronine PLLA+++NA++
Ehsanipour A, et al.,
2021 [120]
ThoracicMouseBDNFHyaluronic acid (HA)-based, spherical microparticle+++NA++
Xie J, et al.,
2022 [121]
ThoracicMouseSonic hedgehog (Shh)
and retinoic acid (RA)
Magnesium oxide (MgO)/
poly (l-lactide-co-ε-caprolactone) (PLCL) scaffold
+++NA+NA
Xi K, et al.,
2020 [122]
ThoracicRatNGFMicroenvironment-responsive
immunoregulatory electrospun fibers
+++NA++
Rooney GE, et al.,
2011 [123]
ThoracicRatDibutyryl cyclic adenosine monophosphate (dbcAMP) Oligo [(polyethylene glycol) fumarate] (OPF) hydrogel scaffoldsNANA+NANANA
Stropkovská A, et al.,
2022 [124]
ThoracicRatRho-A-kinase inhibitorChitosan/collagen porous scaffold +++NA+NA
Man W, et al.,
2021 [72]
ThoracicRatHierarchically aligned fibrin hydrogel Functionalized self-assembling peptides
(fSAP)
++++++
Smith DR, et al.,
2020 [128]
CervicalMouseIL-10 and NT-3 Multiple channel PLG+NA+NA++
Breen BA, et al.,
2017 [130]
ThoracicRatNT-3Injectable collagen scaffold NA++NA++
Wen Y et al.,
2016 [109]
ThoracicRatAntiNogo, BDNF
and vascular endothelial growth factor
Hyaluronic acid (HA) hydrogel ++++++
Jain A, et al.,
2006 [129]
ThoracicRatBDNFGelling agarose hydrogelsNA++NA+NA
Note: +, effective; NA, not available.
Table 5. Summary of the included studies and the effect of combinatory cell therapy for SCI.
Table 5. Summary of the included studies and the effect of combinatory cell therapy for SCI.
Type of Grafted CellsBiomaterial Scaffold Results/AdvantagesLimitations/Disadvantages
NPCs
  • PLGA scaffold
  • HA scaffold
  • Protein-functionalized chitosan scaffold
  • 3D biomimetic hydroge
  • Collagen microchannel scaffold
  • 3D printed heparin sulfate-collagen scaffold
  • Exosomes-collagen scaffold
  • Multi-channel collagen scaffold
  • Aligned collagen scaffold
  • Polymer scaffold
  • Chitosan channels scaffold
  • Protein-functionalized chitosan scaffold
  • Laminin-coated pHEMA-MOETACl Hydrogel
  • Artificial microfiber scaffold
  • Polycaprolactone electrospun fiber scaffold
  • Fibrin scaffold
  • SAP scaffold
  • Matrigel scaffold
  • Functional recovery
  • Graft cells survival and neuronal cell differentiation
  • Secretion of trophic factors
  • Protection of host neuronal cells
  • Axonal outgrowth through injured lesion
  • Remyelination of host axons
  • Neuronal differentiation
  • Host cells survival
  • Immune rejection
  • Tumorgenesis
BMCSs
  • Chitosan-based thermosensitive scaffold
  • Chitosan conduits scaffold
  • Alginate hydrogel biomaterial
  • PLGA scaffold
  • Collagen scaffold
  • Collagen filaments scaffold
  • Porous collagen scaffold
  • NeuroRegen scaffold
  • PLGA scaffold
  • HA-PLL scaffold
  • SAP hydrogel scaffold
  • Biologic scaffolds derived from fibrin and blood plasma
  • Goldnanoparticles (Au NPs)-loaded Agarose/Poly (N-
  • isopropylacrylamide) (PNIPAM)
  • Thermosensitive quaternary ammonium chloride
  • chitosan/β-glycerophosphate (HACC/β-GP) hydrogel scaffold
  • Gelatin sponge scaffold
  • Nanofibrous silk scaffold
  • Cylindrical poly(D,L-lactide-co-glycolide)/small intestinal
  • submucosa scaffold
  • PHEMA scaffold
  • Functional recovery
  • Repair of spinal cord injury
  • Secretion of trophic factors
  • Protection of host neuronal cells
  • Axonal outgrowth
  • Remyelination of host axons
  • Host cells survival
  • Low risk of immune rejection
  • Autologous transplants
  • No ethical issues
  • Difficulty of neuronal differentiation
  • Low cell survival rate
Umbilical MSCs/Wharton’s jelly-derived MSCs
  • Collagen scaffold
  • 3D printed collagen/silk fibroin scaffold
  • HA-PH modified with the integrin-binding peptide
  • arginine-glycine-aspartic acid scaffold
Adipose-derived stem/stromal cells
  • Silk fibroin/chitosan scaffold
  • Matrigel scaffold
  • Resorbable poly(α-hydroxyacid) guidance channels scaffold
Schwann cells (SCs)
  • Biodegradable poly-b-hydroxybutyrate scaffold
  • Oligo[poly(ethylene glycol) fumarate] scaffold
  • Axon growth into SCs implants
  • Ensheathment and myelination
  • No tumorigenicity
  • Modest but significant motor and sensory improvement
  • SCs-elicited responses such as survivability post-transplantation,
  • axon growth, and functional recovery can be improved with
  • appropriate combination treatments
  • Remyelination
  • Functional recovery
  • Secretion of trophic factors
  • No differentiation into
  • neurons and astrocytes
OECs
  • PLGA Scaffold
  • Functional recovery
  • Promotion of cell differentiation
  • Inhibition of astrocyte formation
  • Accelerate neuronal regeneration
  • Secrete nerve growth factors
  • Decrease neuronal apoptosis
  • Reduce glial scaring
  • Produce a number of trophic factors such as VEGF
  • Constitute the myelin and the Ranvier nodes of the axons
  • No differentiation into
  • neurons and astrocytes
Spinal cord-derived ependymal progenitor cells
  • HA containing PLA fibers scaffold
  • Preserve the neuronal tissue
  • Diminish astrocytic reactivity surrounding the scar area
  • Axonal outgrowth
  • No functional recovery
Dental pulp stem cells/Dental follicle cells
  • Chitosan scaffolds
  • Aligned electrospun PCL/PLGA material scaffold
  • Increase the levels of BDNF, GDNF, beta-NGF and NT-3
  • Recovery of hind limb locomotor functions
  • No differentiation into
  • neurons and astrocytes
NPCs and Schwann cells
  • 3D bioprinting of NSC-laden HBC/HA/MA scaffold
  • PLGA scaffolds
  • Biodegradable polymer scaffold
  • Poly (L-lactic-co-glycolic acid) scaffold
  • Provision of an ideal microenvironment for the growth
  • and neural differentiation of grafted cells.
  • Restoration of locomotor function
  • Simulation of the parallel linear structure of spinal cord
  • for optimal neuron regeneration and connection.
  • Immune rejection
  • Tumorgenesis
BMSCs and Schwann cells
  • Multichannel polymer scaffold
  • Functional recovery
  • Secretion of trophic factors
  • Protection of host neuronal cells
  • Axonal outgrowth through injured lesion
  • Remyelination of host axons
  • No differentiation into
  • neurons and astrocytes
Endometrial stem cells and Schwann cells
  • Degradable polymer implant
  • PCL/gelatin nanofibrous scaffold
  • Functional recovery
  • Secretion of trophic factors
  • Protection of host neuronal cells
  • Axonal outgrowth through injured lesion
  • Remyelination of host axons
  • Host cells survival
  • No differentiation into
  • neurons and astrocytes
Dermal fibroblast-reprogrammed neurons
  • 3D silk fibrous material
  • Functional recovery
  • Axonal outgrowth through injured lesion
  • Remyelination of host axons
  • No differentiation into
  • neurons and astrocytes
Adipose-derived stem cells and OECs
  • Serum-derived albumin scaffold
  • Functional recovery
  • Secretion of trophic factors
  • Protection of host neuronal cells
  • Axonal outgrowth through injured lesion
  • Remyelination of host axons
  • No differentiation into
  • neurons and astrocytes
NPCs and MSCs
  • 3D longitudinal scaffold
  • Functional recovery
  • Graft cells survival and neuronal cell differentiation
  • Secretion of trophic factors
  • Protection of host neuronal cells
  • Axonal outgrowth through injured lesion
  • Remyelination of host axons
  • Neuronal differentiation
  • Host cells survival
  • Immune rejection
  • Tumorgenesis
Table 6. Biomaterial scaffolds in clinical trials for SCI (https://www.clinicaltrials.gov/, accessed on 1 August 2022).
Table 6. Biomaterial scaffolds in clinical trials for SCI (https://www.clinicaltrials.gov/, accessed on 1 August 2022).
Effect on Pathophysiological Events
Author, YearLocation of InjuryOngoing Clinical Trials (Identifier)PhaseCombinatory AgentBiomaterial Scaffold Motor FunctionSensory FunctionAnti-InflammationScar/CavityAxon GrowthAngiogenesisFacilitation of Cell Migration
-Cervical/ThoracicNCT02688049Phase 1
Phase 2
NSCs and MSCs NeuroRegen scaffold -------
-Cervical/ThoracicNCT02352077Phase 1Bone marrow mononuclear cells
and MSCs
NeuroRegen scaffold -------
-Cervical/ThoracicNCT02688062Phase 1
Phase 2
Bone marrow mononuclear cells NeuroRegen scaffold -------
-ThoracicNCT02138110Not Applicable Poly(lactic-co-glycolic acid)-b-poly(L-lysine) scaffold-------
-ThoracicNCT03762655Not Applicable Poly(lactic-co-glycolic acid)-b-poly(L-lysine) scaffold-------
-ThoracicNCT02510365Phase 1 Collagen scaffold-------
-Cervical/ThoracicNCT03966794Phase 1
Phase 2
Epidural Electrical StimulationCollagen scaffold-------
-Thoracic/LumbarNCT02326662Phase 1
Phase 2
Autologous NSCsRMx Biomatrix -------
Amr SM, et al.,
2014 [226]
Thoracic--BMSCs/peripheral nerve graftsChitosan-laminin scaffoldSeveral cases improvedSeveral cases improvedNANANANANA
Xiao Z, et al.,
2018 [237]
Cervical/Thoracic--MSCs NeuroRegen scaffold Several cases improvedSeveral cases improvedNANANANANA
Chen W, et al.,
2020 [133]
Thoracic--Bone marrow mononuclear cells NeuroRegen scaffold -Several cases improvedNANANANANA
Note: -, no difference with the control group; NA, not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

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, 2528. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24032528

AMA Style

Suzuki H, Imajo Y, Funaba M, Ikeda H, Nishida N, Sakai T. Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury. International Journal of Molecular Sciences. 2023; 24(3):2528. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24032528

Chicago/Turabian Style

Suzuki, Hidenori, Yasuaki Imajo, Masahiro Funaba, Hiroaki Ikeda, Norihiro Nishida, and Takashi Sakai. 2023. "Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury" International Journal of Molecular Sciences 24, no. 3: 2528. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24032528

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop